101
|
GARRIDO A, MUÑOZ Y, SIERRALTA W, VALLADARES L. Metabolism of Dehydroepiandrosterone Sulfate and Estrone-Sulfate by Human Platelets. Physiol Res 2012; 61:381-8. [DOI: 10.33549/physiolres.932323] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The aim of the present research was to study the uptake of DHEAS, and to establish the intracrine capacity of human platelets to produce sex steroid hormones. The DHEAS transport was evaluated through the uptake of [3H]-DHEAS in the presence or absence of different substrates through the organic anion transporting polypeptide (OATP) family. The activity of sulfatase enzyme was evaluated, and the metabolism of DHEAS was measured by the conversion of [3H]-DHEAS to [3H]-androstenedione, [3H]-testosterone, [3H]-estrone and [3H]-17β-estradiol. Results indicated the existence in the plasma membrane of an OATP with high affinity for DHEAS and estrone sulphate (E1S). The platelets showed the capacity to convert DHEAS to active DHEA by the steroid-sulfatase activity. The cells resulted to be a potential site for androgens production, since they have the capacity to produce androstenedione and testosterone; in addition, they reduced [3H]-estrone to [3H]-17β-estradiol. This is the first demonstration that human platelets are able to import DHEAS and E1S using the OATP family and to convert DHEAS to active DHEA, and to transform E1S to 17β-estradiol.
Collapse
Affiliation(s)
| | | | | | - L. VALLADARES
- Institute of Nutrition and Food Technology, Universidad de Chile, Santiago, Chile
| |
Collapse
|
102
|
Abstract
The platelet release reaction plays a critical role in thrombosis and contributes to the events that follow hemostasis. Previous studies have shown that platelet secretion is mediated by Soluble NSF Attachment Protein Receptor (SNARE) proteins from granule and plasma membranes. The SNAREs form transmembrane complexes that mediate membrane fusion and granule cargo release. Although VAMP-8 (v-SNARE) and SNAP-23 (a t-SNARE class) are important for platelet secretion, the identity of the functional syntaxin (another t-SNARE class) has been controversial. Previous studies using anti-syntaxin Abs in permeabilized platelets have suggested roles for both syntaxin-2 and syntaxin-4. In the present study, we tested these conclusions using platelets from syntaxin-knockout mouse strains and from a Familial Hemophagocytic Lymphohistiocytosis type 4 (FHL4) patient. Platelets from syntaxin-2 and syntaxin-4 single- or double-knockout mice had no secretion defect. Platelets from a FHL4 patient deficient in syntaxin-11 had a robust defect in agonist-induced secretion although their morphology, activation, and cargo levels appeared normal. Semiquantitative Western blotting showed that syntaxin-11 is the more abundant syntaxin in both human and murine platelets. Coimmunoprecipitation experiments showed that syntaxin-11 can form SNARE complexes with both VAMP-8 and SNAP-23. The results of the present study indicate that syntaxin-11, but not syntaxin-2 or syntaxin-4, is required for platelet exocytosis.
Collapse
|
103
|
Abstract
The role of platelets in hemostasis and thrombosis is clearly established; however, the mechanisms by which platelets mediate inflammatory and immune pathways are less well understood. Platelets interact and modulate the function of blood and vascular cells by releasing bioactive molecules. Although the platelet is anucleate, it contains transcripts that may mirror disease. Platelet mRNA is only associated with low-level protein translation; however, platelets have a unique membrane structure allowing for the passage of small molecules, leading to the possibility that its cytoplasmic RNA may be passed to nucleated cells. To examine this question, platelet-like particles with labeled RNA were cocultured with vascular cells. Coculture of platelet-like particles with activated THP-1, monocytic, and endothelial cells led to visual and functional RNA transfer. Posttransfer microarray gene expression analysis of THP-1 cells showed an increase in HBG1/HBG2 and HBA1/HBA2 expression that was directly related to the transfer. Infusion of wild-type platelets into a TLR2-deficient mouse model established in vivo confirmation of select platelet RNA transfer to leukocytes. By specifically transferring green fluorescent protein, we also observed external RNA was functional in the recipient cells. The observation that platelets possess the capacity to transfer cytosolic RNA suggests a new function for platelets in the regulation of vascular homeostasis.
Collapse
|
104
|
Off-Target Platelet Activation in Macaques Unique to a Therapeutic Monoclonal Antibody. Toxicol Pathol 2012; 40:899-917. [DOI: 10.1177/0192623312444029] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
AMG X, a human neutralizing monoclonal antibody (mAb) against a soluble human protein, caused thrombocytopenia, platelet activation, reduced mean arterial pressure, and transient loss of consciousness in cynomolgus monkeys after first intravenous administration. In vitro, AMG X induced activation in platelets from macaque species but not from humans or baboons. Other similar mAbs against the same pharmacological target failed to induce these in vivo and in vitro effects. In addition, the target protein was known to not be expressed on platelets, suggesting that platelet activation occurred through an off-target mechanism. AMG X bound directly to cynomolgus platelets and required both the Fab and Fc portion of the mAb for platelet activation. Binding to platelets was inhibited by preincubation of AMG X with its pharmacological target or with anti-human Fc antibodies or by preincubation of platelets with AMG X F(ab′)2 or human immunoglobulin (IVIG). AMG X F(ab′)2 did not activate platelets. Thus, platelet activation required both recognition/binding of a platelet ligand with the Fab domain and interaction of platelet Fc receptors (i.e., FcγRIIa) with the Fc domain. These findings reflect the complexity of the mechanism of action of mAbs and the increasing awareness of potential for unintended effects in preclinical species.
Collapse
|
105
|
Reilly SJ, Li N, Liska J, Ekström M, Tornvall P. Coronary artery bypass graft surgery up-regulates genes involved in platelet aggregation. J Thromb Haemost 2012; 10:557-63. [PMID: 22329762 DOI: 10.1111/j.1538-7836.2012.04660.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND During and shortly after coronary artery bypass graft (CABG) surgery, there is an increase in thromboembolic events. CABG, a strong inflammatory stimulus, is associated with a hypercoaguable state. Platelets might contribute to this hypercoaguable state because they have a pivotal role in thrombosis. In the days following surgery there is augmented platelet regeneration in response to the inflammatory stimulus. OBJECTIVES The aim of this study was to investigate any changes in platelet mRNA profiles to test the hypothesis that post-CABG surgery platelets are associated with a prothrombotic state. METHODS Blood was sampled and platelets purified from 11 patients before and 3-6 days after CABG. Gene expression profiling was performed using low density array (LDA) plates for seven of the patients. RESULTS Forty-five genes were examined and those significantly up-regulated were glycoprotein (GP)IIb, GPIIIa and cyclooxygenase-1 (COX-1). These findings were confirmed in four more patients, including flow cytometry analysis of the GPIIb/IIIa receptor. CONCLUSIONS CABG surgery up-regulates mRNA and protein levels of proteins that are key players in platelet aggregation. Marked elevation of GPIIb/IIIa mRNA levels results in significantly increased GPIIb/IIIa expression in platelets post-CABG surgery, which may be a reason for increased thrombus formation and myocardial infarction after CABG.
Collapse
Affiliation(s)
- S-J Reilly
- Department of Medicine, Atherosclerosis Research Unit, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | | |
Collapse
|
106
|
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
|
107
|
Amisten S. A rapid and efficient platelet purification protocol for platelet gene expression studies. Methods Mol Biol 2012; 788:155-172. [PMID: 22130707 DOI: 10.1007/978-1-61779-307-3_12] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Isolation of pure platelet samples from whole blood is crucial for the study of platelet gene expression. The main obstacles to overcome in order to successfully isolate platelets from whole blood include (1) platelet activation; (2) leukocyte and red blood cell contamination, and (3) time-dependent platelet mRNA degradation. This chapter describes a rapid and highly efficient method for isolating human circulating platelets from small volumes of whole blood based on efficient inhibition of platelet activation and leukocyte removal by filtration followed by magnetic bead-depletion of residual contaminating leukocytes and red blood cells. Also described are methods for RNA extraction, cDNA synthesis, and platelet gene expression studies using both quantitative real-time PCR and microarray.
Collapse
Affiliation(s)
- Stefan Amisten
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, Oxford University, The Churchill Hospital, Oxford, UK.
| |
Collapse
|
108
|
Abstract
Although platelets are the smallest cells in the blood, they are implied in various processes ranging from immunology and oncology to thrombosis and hemostasis. Many large-scale screening programs, genome-wide association, and "omics" studies have generated lists of genes and loci that are probably involved in the formation or physiology of platelets under normal and pathologic conditions. This creates an increasing demand for new and improved model systems that allow functional assessment of the corresponding gene products in vivo. Such animal models not only render invaluable insight in the platelet biology, but in addition, provide improved test systems for the validation of newly developed anti-thrombotics. This review summarizes the most important models to generate transgenic platelets and to study their influence on platelet physiology in vivo. Here we focus on the zebrafish morpholino oligonucleotide technology, the (platelet-specific) knockout mouse, and the transplantation of genetically modified human or murine platelet progenitor cells in myelo-conditioned mice. The various strengths and pitfalls of these animal models are illustrated by recent examples from the platelet field. Finally, we highlight the latest developments in genetic engineering techniques and their possible application in platelet research.
Collapse
|
109
|
Eskan MA, Greenwell H. Theoretical and Clinical Considerations for Autologous Blood Preparations: Platelet-Rich Plasma, Fibrin Sealants, and Plasma-Rich Growth Factors. Clin Adv Periodontics 2011. [DOI: 10.1902/cap.2011.110034] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
110
|
The PPAR-Platelet Connection: Modulators of Inflammation and Potential Cardiovascular Effects. PPAR Res 2011; 2008:328172. [PMID: 18288284 PMCID: PMC2233896 DOI: 10.1155/2008/328172] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2007] [Accepted: 11/06/2007] [Indexed: 01/08/2023] Open
Abstract
Historically, platelets were viewed as simple anucleate cells responsible for initiating thrombosis and maintaining
hemostasis, but clearly they are also key mediators of inflammation and immune cell activation. An emerging body of
evidence links platelet function and thrombosis to vascular inflammation. peroxisome proliferator-activated receptors
(PPARs) play a major role in modulating inflammation and, interestingly, PPARs (PPARβ/δ and PPARγ) were recently
identified in platelets. Additionally, PPAR agonists attenuate platelet activation; an important discovery for two reasons.
First, activated platelets are formidable antagonists that initiate and prolong a cascade of events that contribute to
cardiovascular disease (CVD) progression. Dampening platelet release of proinflammatory mediators, including
CD40 ligand (CD40L, CD154), is essential to hinder this cascade. Second, understanding the biologic importance
of platelet PPARs and the mechanism(s) by which PPARs regulate platelet activation will be imperative in designing
therapeutic strategies lacking the deleterious or unwanted side effects of current treatment options.
Collapse
|
111
|
Lordkipanidzé M, Pharand C, Schampaert E, Palisaitis DA, Diodati JG. Heterogeneity in platelet cyclooxygenase inhibition by aspirin in coronary artery disease. Int J Cardiol 2011; 150:39-44. [DOI: 10.1016/j.ijcard.2010.02.025] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2009] [Revised: 01/12/2010] [Accepted: 02/13/2010] [Indexed: 11/29/2022]
|
112
|
Calverley DC, Phang TL, Choudhury QG, Gao B, Oton AB, Weyant MJ, Geraci MW. Significant downregulation of platelet gene expression in metastatic lung cancer. Clin Transl Sci 2011; 3:227-32. [PMID: 21500395 DOI: 10.1111/j.1752-8062.2010.00226.x] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Platelets play a major role in the metastatic dissemination of tumor cells in vivo . Recent evidence reveals megakaryocyte-derived platelet pre-mRNA is spliced to mRNA and then translated into functional proteins in response to external stimulation. Employing a human lung cancer model, we hypothesized a subset of megakaryocyte/platelet genes exists that are significantly over or underexpressed in metastasis compared with noncancer. Microarray analysis employing platelet mRNA followed by unsupervised hierarchical clustering revealed an expression profile that includes decreased expression of 197 of the 200 platelet genes with the most altered expression (p < 1.0 × 10(-4)). Among the 608 splicing events identified between the metastasis and negative control groups, 33 highly variable genes were identified with between 3 and 13 splicing events each. In conclusion, this preliminary study reveals a platelet-based gene expression signature that differentiates metastatic lung cancer from negative controls on the basis of decreased expression of 197 of the 200 genes with the most altered expression levels. Further study may yield a prognostic tool for future metastasis among subsets of early stage lung cancer patients.
Collapse
|
113
|
Gosk-Bierska I, McBane RD, Wu Y, Mruk J, Tafur A, McLeod T, Wysokinski WE. Platelet factor XIII gene expression and embolic propensity in atrial fibrillation. Thromb Haemost 2011; 106:75-82. [PMID: 21655673 DOI: 10.1160/th10-11-0765] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2010] [Accepted: 04/25/2011] [Indexed: 12/12/2022]
Abstract
Nearly 15% of patients with non-valvular atrial fibrillation (NVAF) have left atrial appendage thrombus (LAAT) by transesophageal echocardiography (TEE) and yet the annual stroke rate averages 5%. The aim of this study was to identify variables influencing embolic propensity of LAAT. Platelet RNA was extracted from platelet-rich regions within formalin-fixed, paraffin-embedded specimens obtained from NVAF patients during cardiac surgery (26 LAAT from 23 patients) or peripheral embolectomy (51 thrombi from 41 patients). Platelet RNA was also assessed from whole blood from 40 NVAF patients. Expression of six platelet-predominate genes: H2A histone family, A1 domain of factor XIII, integrin α₂bβ₃; glycoprotein IX, platelet factor 4, glycoprotein Ib, was performed using TaqMan MGB-probe based quantitative real-time polymerase chain reaction. Platelet factor XIII subunit A gene expression was significantly lower in embolised compared to non-embolised thrombi as determined by normalised cycle threshold values (4.0 ± 1.2 v 2.8 ± 1.8, p=0.02). Expression of other genes did not differ by embolic status. In conclusion, RNA extracted from formalin-fixed, paraffin-embedded platelet-rich tissues can be used for analysis of platelet-predominate gene expression. Variable factor XIII gene expression in thrombi generated during NVAF may in part explain the propensity to embolisation.
Collapse
|
114
|
Shah NA, Hills NK, Waleh N, McCurnin D, Seidner S, Chemtob S, Clyman R. Relationship between circulating platelet counts and ductus arteriosus patency after indomethacin treatment. J Pediatr 2011; 158:919-923.e1-2. [PMID: 21195414 PMCID: PMC3095765 DOI: 10.1016/j.jpeds.2010.11.018] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2010] [Revised: 10/18/2010] [Accepted: 11/05/2010] [Indexed: 11/28/2022]
Abstract
OBJECTIVE To determine whether low platelet counts are related to the incidence of patent ductus arteriosus (PDA) after indomethacin treatment in preterm human infants. STUDY DESIGN Multivariable logistic regression modeling was used for a cohort of 497 infants, who received indomethacin (within 15 hours of birth). RESULTS Platelet counts were not related to the incidence of permanent closure after indomethacin constriction. There was a relationship between platelet counts and the initial degree of constriction; however, this relationship appeared to be primarily influenced by the high end of the platelet distribution curve. PDA incidence was similar in infants with platelet counts < 50 × 10⁹/L and those with platelet counts above this range. Only when platelet counts were consistently >230 ×10⁹/L was there a decrease in PDA incidence. CONCLUSION In contrast to the evidence in mice, low circulating platelet counts do not affect permanent ductus closure (or ductus reopening) in human preterm infants.
Collapse
Affiliation(s)
- Nidhi A. Shah
- Department of Pediatrics, University of California San Francisco, CA 94143
| | - Nancy K. Hills
- Department of Neurology, University of California San Francisco, CA 94143
| | - Nahid Waleh
- Pharmaceutical Discovery Division, SRI International, Menlo Park, CA 94025
| | - Donald McCurnin
- Department of Pediatrics, University of Texas, Health Science Center, San Antonio, TX, 78229, Southwest Foundation for Biomedical Research, San Antonio TX, 78227
| | - Steven Seidner
- Department of Pediatrics, University of Texas, Health Science Center, San Antonio, TX, 78229, Southwest Foundation for Biomedical Research, San Antonio TX, 78227
| | - Sylvain Chemtob
- Departments of Pediatrics, Ophthalmology and Pharmacology Research Center, Hôpital Ste. Justine, Montreal, Quebec, H3T-1C5, Canada
| | - Ronald Clyman
- Department of Pediatrics, University of California San Francisco, CA 94143, Cardiovascular Research Institute, University of California San Francisco, CA 94143
| |
Collapse
|
115
|
Megakaryocytes differentially sort mRNAs for matrix metalloproteinases and their inhibitors into platelets: a mechanism for regulating synthetic events. Blood 2011; 118:1903-11. [PMID: 21628401 DOI: 10.1182/blood-2010-12-324517] [Citation(s) in RCA: 117] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Megakaryocytes transfer a diverse and functional transcriptome to platelets during the final stages of thrombopoiesis. In platelets, these transcripts reflect the expression of their corresponding proteins and, in some cases, serve as a template for translation. It is not known, however, if megakaryocytes differentially sort mRNAs into platelets. Given their critical role in vascular remodeling and inflammation, we determined whether megakaryocytes selectively dispense transcripts for matrix metalloproteinases (MMPs) and their tissue inhibitors (TIMPs) into platelets. Next-generation sequencing (RNA-Seq) revealed that megakaryocytes express mRNA for 10 of the 24 human MMP family members. mRNA for all of these MMPs are present in platelets with the exception of MMP-2, 14, and 15. Megakaryocytes and platelets also express mRNA for TIMPs 1-3, but not TIMP-4. mRNA expression patterns predicted the presence and, in most cases, the abundance of each corresponding protein. Nonetheless, exceptions were observed: MMP-2 protein is present in platelets but not its transcript. In contrast, quiescent platelets express TIMP-2 mRNA but only traces of TIMP-2 protein. In response to activating signals, however, platelets synthesize significant amounts of TIMP-2 protein. These results demonstrate that megakaryocytes differentially express mRNAs for MMPs and TIMPs and selectively transfer a subset of these into platelets. Among the platelet messages, TIMP-2 serves as a template for signal-dependent translation.
Collapse
|
116
|
Gnatenko DV, Bahou WF. The Application of Microarray Analysis and its Integration with Proteomics for Study of Platelet‐Associated Disorders. PLATELET PROTEOMICS 2011:231-251. [DOI: 10.1002/9780470940297.ch10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
117
|
Fei SS, Wilmarth PA, Hitzemann RJ, McWeeney SK, Belknap JK, David LL. Protein database and quantitative analysis considerations when integrating genetics and proteomics to compare mouse strains. J Proteome Res 2011; 10:2905-12. [PMID: 21553863 PMCID: PMC3128464 DOI: 10.1021/pr200133p] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Decades of genetics research comparing mouse strains has identified many regions of the genome associated with quantitative traits. Microarrays have been used to identify which genes in those regions are differentially expressed and are therefore potentially causal; however, genetic variants that affect probe hybridization lead to many false conclusions. Here we used spectral counting to compare brain striata between two mouse strains. Using strain-specific protein databases, we concluded that proteomics was more robust to sequence differences than microarrays; however, some proteins were still significantly affected. To generate strain-specific databases, we used a complete database that contained all of the putative genetic isoforms for each protein. While the increased proteome coverage in the databases led to a 6.8% gain in peptide assignments compared to a nonredundant database, it also necessitated the development of a strategy for grouping similar proteins due to a large number of shared peptides. Of the 4563 identified proteins (2.1% FDR), there were 1807 quantifiable proteins/groups that exceeded minimum count cutoffs. With four pooled biological replicates per strain, we used quantile normalization, ComBat (a package that adjusts for batch effects), and edgeR (a package for differential expression analysis of count data) to identify 101 differentially expressed proteins/groups, 84 of which had a coding region within one of the genomic regions of interest identified by the Portland Alcohol Research Center.
Collapse
Affiliation(s)
- Suzanne S Fei
- Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, Oregon 97239, USA.
| | | | | | | | | | | |
Collapse
|
118
|
Colombo G, Gertow K, Marenzi G, Brambilla M, De Metrio M, Tremoli E, Camera M. Gene expression profiling reveals multiple differences in platelets from patients with stable angina or non-ST elevation acute coronary syndrome. Thromb Res 2011; 128:161-8. [PMID: 21420725 DOI: 10.1016/j.thromres.2011.02.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2010] [Revised: 02/16/2011] [Accepted: 02/21/2011] [Indexed: 11/26/2022]
Abstract
INTRODUCTION Platelets play a key role in coronary artery disease. They have the capacity of protein synthesis through translation of megakaryocyte-derived mRNAs, which may influence pathophysiological functions. The present study aimed to prove the concept that platelets from patients with non-ST elevation acute coronary syndrome (NSTE-ACS) have differential mRNA expression profiles, in the hypothesis that this may influence their thrombogenicity. MATERIALS AND METHODS Gene expression profiles were determined in RNA pools from resting platelets of patients with stable angina (SA, n = 14) or NSTE-ACS (n = 15) using a glass microarray platform. Validation was done by real-time PCR and immunoblot analyses in independent sets of individual samples (26 SA and 17 NSTE-ACS patients, in total). Parallel comparison with healthy subjects was performed to relate the relative abundance of validated genes in CAD patients to a control expression level. RESULTS Microarray analysis identified 45 transcripts with a significant ≥ ± 2.0-fold difference in expression between NSTE-ACS and SA platelet pools. Thus, gene expression profiles at least partially discriminate unstable from stable CAD. Validation confirmed a significant over-expression of 3 genes in NSTE-ACS at both mRNA and protein level. In particular, the glycoprotein Ib β-polypeptide (GP1BB) was increased in NSTE-ACS also in comparison with healthy subjects. CONCLUSION This study provides evidence that NSTE-ACS platelets are potentially preconditioned to a higher degree of reactivity on the transcriptional level. Our data suggest that a different composition of the mRNA pool might mediate an increased platelet prothrombotic potential in NSTE-ACS patients.
Collapse
|
119
|
|
120
|
Abstract
Platelets are specialized blood cells that play central roles in physiologic and pathologic processes of hemostasis, wound healing, host defense, thrombosis, inflammation, and tumor metastasis. Activation of platelets is crucial for platelet function that includes a complex interplay of adhesion, signaling molecules, and release of bioactive factors. Transfusion of platelet concentrates is an important treatment component for thrombocytopenia and bleeding. Recent progress in high-throughput mRNA and protein profiling techniques has advanced the understanding of platelet biological functions toward identifying novel platelet-expressed and secreted proteins, analyzing functional changes between normal and pathologic states, and determining the effects of processing and storage on platelet concentrates for transfusion. It is important to understand the different standard methods of platelet preparation and how they differ from the perspective for use as research samples in clinical chemistry. Two simple methods are described here for the preparation of research-scale platelet samples from whole blood, and detailed notes are provided about the methods used for the preparation of platelet concentrates for transfusion.
Collapse
|
121
|
Abstract
Sudden cardiac death (SCD) remains one of the most prevalent modes of death in industrialized countries, and myocardial ischemia due to thrombotic coronary occlusion is its primary cause. The role of platelets in the occurrence of SCD extends beyond coronary flow impairment by clot formation. Here we review the substances released by platelets during clot formation and their arrhythmic properties. Platelet products are released from three types of platelet granules: dense core granules, alpha-granules, and platelet lysosomes. The physiologic properties of dense granule products are of special interest as a potential source of arrhythmic substances. They are released readily upon activation and contain high concentrations of serotonin, histamine, purines, pyrimidines, and ions such as calcium and magnesium. Potential arrhythmic mechanisms of these substances, e.g., serotonin and high energy phosphates, include induction of coronary constriction, calcium overloading, and induction of delayed after-depolarizations. Alpha-granules produce thromboxanes and other arachidonic-acid products with many potential arrhythmic effects mediated by interference with cardiac sodium, calcium, and potassium channels. Alpha-granules also contain hundreds of proteins that could potentially serve as ligands to receptors on cardiomyocytes. Lysosomal products probably do not have an important arrhythmic effect. Platelet products and ischemia can induce coronary permeability, thereby enhancing interaction with surrounding cardiomyocytes. Antiplatelet therapy is known to improve survival after myocardial infarction. Although an important part of this effect results from prevention of coronary clot formation, there is evidence to suggest that antiplatelet therapy also induces anti-arrhythmic effects during ischemia by preventing the release of platelet activation products.
Collapse
|
122
|
Williams MS, Weiss EJ, Sabatine MS, Simon DI, Bahou WF, Becker LC, Parise LV, Dauerman HL, French PA, Smyth SS, Becker RC. Genetic regulation of platelet receptor expression and function: application in clinical practice and drug development. Arterioscler Thromb Vasc Biol 2010; 30:2372-84. [PMID: 21084706 DOI: 10.1161/atvbaha.110.218131] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Understanding genetic contributions to platelet function could have profound clinical ramifications for personalizing platelet-directed pharmacotherapy, by providing insight into the risks and possible benefits associated with specific genotypes. This article represents an integrated summary of presentations related to genetic regulation of platelet receptor expression and function given at the Fifth Annual Platelet Colloquium in January 2010. It is supplemented with additional highlights from the literature covering (1) approaches to determining and evidence for the associations of genetic variants with platelet hypo- and hyperresponsive phenotypes, (2) the ramifications of these polymorphisms with regard to clinical responses to antiplatelet therapies, and (3) the role of platelet function/genetic testing in guiding antiplatelet therapy.
Collapse
Affiliation(s)
- Marlene S Williams
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Md 21224, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
123
|
Liumbruno G, D’Alessandro A, Grazzini G, Zolla L. How has proteomics informed transfusion biology so far? Crit Rev Oncol Hematol 2010; 76:153-72. [DOI: 10.1016/j.critrevonc.2010.01.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2009] [Revised: 12/22/2009] [Accepted: 01/07/2010] [Indexed: 01/06/2023] Open
|
124
|
Spinelli SL, Maggirwar SB, Blumberg N, Phipps RP. Nuclear emancipation: a platelet tour de force. Sci Signal 2010; 3:pe37. [PMID: 20959522 DOI: 10.1126/scisignal.3144pe37] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Mammalian platelets are anucleate cells produced by the polyploid megakaryocyte. Platelets are more than just key players in hemostasis (blood clotting in response to injury); they also have important roles in inflammation, immunity, tumor progression, and thrombosis. Complex systems of homeostasis have been described for platelets, including posttranscriptional and translational mechanisms to regulate platelet function. Platelets contain transcription factors, and these proteins have essential roles in regulating nongenomic processes. A study provides evidence for a previously unknown negative feedback pathway for limiting platelet activation that occurs through the nuclear factor κB transcription factor family. This pathway is mediated by an adenosine 3',5'-monophosphate-independent protein kinase A activity in response to platelet stimulation. Our appreciation of the role of transcription factors in mammalian platelet biology is nascent but holds great promise for both understanding platelet function and translation into clinical uses.
Collapse
Affiliation(s)
- Sherry L Spinelli
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | | | | | | |
Collapse
|
125
|
Proteomic evaluation on antiplatelet activity of piperlongumine derived from Piper longum. Mol Cell Toxicol 2010. [DOI: 10.1007/s13273-010-0040-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
126
|
Calverley DC, Casserly IP, Choudhury QG, Phang TL, Gao B, Messenger JC, Geraci MW. Platelet Gene Expression as a Biomarker Risk Stratification Tool in Acute Myocardial Infarction: A Pilot Investigation. Gulf J Oncolog 2010. [DOI: 10.4137/cmbd.s5005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Platelets play a major role in the pathophysiology of acute myocardial infarction (AMI). Recent evidence reveals megakaryocyte-derived platelet pre-mRNA is spliced to mRNA and then translated into functional proteins in response to external stimulation. An exon microarray analyzes pre-mRNA alternative splicing and is thus applicable for studying gene expression in the anucleate platelet. We hypothesized a subset of megakaryocyte/platelet genes exists that are significantly over or underexpressed in AMI compared with stable coronary artery disease (CAD), yielding a gene expression profile for further study. Microarray analysis employing platelet mRNA was used to generate gene expression data in the above two patient groups. Unsupervised hierarchical clustering has revealed an expression profile that includes 95 over- or under-expressed genes depicted in a heat map where separation of both sets takes place. This preliminary study reveals a platelet-based gene expression signature that differentiates between AMI and stable CAD, and further study may yield a prognostic tool for a future AMI event in atherosclerosis risk factor-based subsets of CAD patients.
Collapse
Affiliation(s)
- David C. Calverley
- Divisions of Hematology, Department of Medicine, University of Colorado, Denver, CO 80045, USA
| | - Ivan P. Casserly
- Cardiology, Department of Medicine, University of Colorado, Denver, CO 80045, USA
| | - Qamrul G. Choudhury
- Divisions of Hematology, Department of Medicine, University of Colorado, Denver, CO 80045, USA
| | - Tzu L. Phang
- Pulmonary sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Denver, CO 80045, USA
| | - Bifeng Gao
- Pulmonary sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Denver, CO 80045, USA
| | - John C. Messenger
- Cardiology, Department of Medicine, University of Colorado, Denver, CO 80045, USA
| | - Mark W. Geraci
- Pulmonary sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Denver, CO 80045, USA
| |
Collapse
|
127
|
Production and new perspectives of a blood transfusion service at the Brazilian National Cancer Institute in Rio De Janeiro, Brazil. BLOOD TRANSFUSION = TRASFUSIONE DEL SANGUE 2010; 8 Suppl 3:s82-5. [PMID: 20606755 DOI: 10.2450/2010.013s] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
128
|
Proteomics strategy for identifying candidate bioactive proteins in complex mixtures: application to the platelet releasate. J Biomed Biotechnol 2010; 2010:107859. [PMID: 20368775 PMCID: PMC2846341 DOI: 10.1155/2010/107859] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2009] [Revised: 10/06/2009] [Accepted: 11/10/2009] [Indexed: 02/08/2023] Open
Abstract
Proteomic approaches have proven powerful at identifying large numbers of proteins, but there are fewer reports of functional characterization of proteins in biological tissues. Here, we describe an experimental approach that fractionates proteins released from human platelets, linking bioassay activity to identity. We used consecutive orthogonal separation platforms to ensure sensitive detection: (a) ion-exchange of intact proteins, (b) SDS-PAGE separation of ion-exchange fractions and (c) HPLC separation of tryptic digests coupled to electrospray tandem mass spectrometry. Migration of THP-1 monocytes in response to complete or fractionated platelet releasate was assessed and located to just one of the forty-nine ion-exchange fractions. Over 300 proteins were identified in the releasate, with a wide range of annotated biophysical and biochemical properties, in particular platelet activation, adhesion, and wound healing. The presence of PEDF and involucrin, two proteins not previously reported in platelet releasate, was confirmed by western blotting. Proteins identified within the fraction with monocyte promigratory activity and not in other inactive fractions included vimentin, PEDF, and TIMP-1. We conclude that this analytical platform is effective for the characterization of complex bioactive samples.
Collapse
|
129
|
Schubert P, Devine DV. De novo protein synthesis in mature platelets: a consideration for transfusion medicine. Vox Sang 2010; 99:112-22. [PMID: 20345520 DOI: 10.1111/j.1423-0410.2010.01333.x] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Platelet function in thrombosis and haemostasis is reasonably well understood at the molecular level with respect to the proteins involved in cellular structure, signalling networks and platelet interaction with clotting factors and other cells. However, the natural history of these proteins has only recently garnered the attention of platelet researchers. De novo protein synthesis in platelets was discovered 40 years ago; however, it was generally dismissed as merely an interesting minor phenomenon until studies over the past few years renewed interest in this aspect of platelet proteins. It is now accepted that anucleate platelets not only have the potential to synthesize proteins, but this capacity seems to be required to fulfil their function. With translational control as the primary mode of regulation, platelets are able to express biologically relevant gene products in a timely and signal-dependent manner. Platelet protein synthesis during storage of platelet concentrates is a nascent area of research. Protein synthesis does occur, although not for all proteins found in the platelet protein profile. Furthermore, mRNA appears to be well preserved under standard storage conditions. Although its significance is not yet understood, the ability to replace proteins may form a type of cellular repair mechanism during storage. Disruption by inappropriate storage conditions or processes that block protein synthesis such as pathogen reduction technologies may have direct effects on the ability of platelets to synthesize proteins during storage.
Collapse
Affiliation(s)
- P Schubert
- Canadian Blood Services and the Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
| | | |
Collapse
|
130
|
RUNX1/core binding factor A2 regulates platelet 12-lipoxygenase gene (ALOX12): studies in human RUNX1 haplodeficiency. Blood 2010; 115:3128-35. [PMID: 20181616 DOI: 10.1182/blood-2009-04-214601] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Haploinsufficiency of RUNX1 (also known as CBFA2/AML1) is associated with familial thrombocytopenia, platelet dysfunction, and predisposition to acute leukemia. We have reported on a patient with thrombocytopenia and impaired agonist-induced aggregation, secretion, and protein phosphorylation associated with a RUNX1 mutation. Expression profiling of platelets revealed approximately 5-fold decreased expression of 12-lipoxygenase (12-LO, gene ALOX12), which catalyzes 12-hydroxyeicosatetraenoic acid production from arachidonic acid. We hypothesized that ALOX12 is a direct transcriptional target gene of RUNX1. In present studies, agonist-induced platelet 12-HETE production was decreased in the patient. Four RUNX1 consensus sites were identified in the 2-kb promoter region of ALOX12 (at -1498, -1491, -708, -526 from ATG). In luciferase reporter studies in human erythroleukemia cells, mutation of each site decreased activity; overexpression of RUNX1 up-regulated promoter activity, which was abolished by mutation of RUNX1 sites. Gel shift studies, including with recombinant protein, revealed RUNX1 binding to each site. Chromatin immunoprecipitation revealed in vivo RUNX1 binding in the region of interest. siRNA knockdown of RUNX1 decreased RUNX1 and 12-LO proteins. ALOX12 is a direct transcriptional target of RUNX1. Our studies provide further proof of principle that platelet expression profiling can elucidate novel alterations in platelets with inherited dysfunction.
Collapse
|
131
|
Horstman LL, Jy W, Ahn YS, Zivadinov R, Maghzi AH, Etemadifar M, Steven Alexander J, Minagar A. Role of platelets in neuroinflammation: a wide-angle perspective. J Neuroinflammation 2010; 7:10. [PMID: 20128908 PMCID: PMC2829540 DOI: 10.1186/1742-2094-7-10] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2009] [Accepted: 02/03/2010] [Indexed: 01/03/2023] Open
Abstract
OBJECTIVES This review summarizes recent developments in platelet biology relevant to neuroinflammatory disorders. Multiple sclerosis (MS) is taken as the "Poster Child" of these disorders but the implications are wide. The role of platelets in inflammation is well appreciated in the cardiovascular and cancer research communities but appears to be relatively neglected in neurological research. ORGANIZATION After a brief introduction to platelets, topics covered include the matrix metalloproteinases, platelet chemokines, cytokines and growth factors, the recent finding of platelet PPAR receptors and Toll-like receptors, complement, bioactive lipids, and other agents/functions likely to be relevant in neuroinflammatory diseases. Each section cites literature linking the topic to areas of active research in MS or other disorders, including especially Alzheimer's disease. CONCLUSION The final section summarizes evidence of platelet involvement in MS. The general conclusion is that platelets may be key players in MS and related disorders, and warrant more attention in neurological research.
Collapse
Affiliation(s)
- Lawrence L Horstman
- Wallace Coulter Platelet Laboratory, Division of Hematology and Oncology, Department of Medicine, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Wenche Jy
- Wallace Coulter Platelet Laboratory, Division of Hematology and Oncology, Department of Medicine, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Yeon S Ahn
- Wallace Coulter Platelet Laboratory, Division of Hematology and Oncology, Department of Medicine, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Robert Zivadinov
- Buffalo Neuroimaging Analysis Center, The Jacobs Neurological Institute, Department of Neurology, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo NY, USA
| | - Amir H Maghzi
- Department of Neurology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Masoud Etemadifar
- Department of Neurology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - J Steven Alexander
- Department of Cellular and Molecular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA
| | - Alireza Minagar
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA
| |
Collapse
|
132
|
Abstract
Many bacteria are capable of interacting with platelets and inducing platelet aggregation. This interaction may be a direct interaction between a bacterial surface protein and a platelet receptor or may be an indirect interaction where plasma proteins bind to the bacterial surface and subsequently bind to a platelet receptor. However, these interactions usually do not trigger platelet activation as a secondary co-signal is also required. This is usually due to specific antibody bound to the bacteria interacting with FcgammaRIIa on the platelet surface. Secreted bacterial products such as gingipains and lipopolysaccharide may also be capable of triggering platelet activation.
Collapse
Affiliation(s)
- Steven W. Kerrigan
- School of Pharmacy, Royal College of Surgeons in Ireland, 123 St Stephens Green, Dublin 2, Ireland
- Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, 123 St Stephens Green, Dublin 2, Ireland
| | - Dermot Cox
- Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, 123 St Stephens Green, Dublin 2, Ireland
| |
Collapse
|
133
|
Kondkar AA, Bray MS, Leal SM, Nagalla S, Liu DJ, Jin Y, Dong JF, Ren Q, Whiteheart SW, Shaw C, Bray PF. VAMP8/endobrevin is overexpressed in hyperreactive human platelets: suggested role for platelet microRNA. J Thromb Haemost 2010; 8:369-78. [PMID: 19943878 PMCID: PMC3312605 DOI: 10.1111/j.1538-7836.2009.03700.x] [Citation(s) in RCA: 149] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND Variation in platelet reactivity contributes to disorders of hemostasis and thrombosis, but the molecular mechanisms are not well understood. OBJECTIVES To discover associations between interindividual platelet variability and the responsible platelet genes, and to begin to define the molecular mechanisms altering platelet gene expression. SUBJECTS/METHODS Two hundred and eighty-eight healthy subjects were phenotyped for platelet responsiveness. Platelet RNA from subjects demonstrating hyperreactivity (n=18) and hyporeactivity (n=11) was used to screen the human transcriptome. RESULTS Distinctly different mRNA profiles were observed between subjects with differing platelet reactivity. Increased levels of mRNA for VAMP8/endobrevin, a critical v-SNARE involved in platelet granule secretion, were associated with platelet hyperreactivity (Q=0.0275). Validation studies of microarray results showed 4.8-fold higher mean VAMP8 mRNA levels in hyperreactive than hyporeactive platelets (P=0.0023). VAMP8 protein levels varied 13-fold among platelets from these normal subjects, and were 2.5-fold higher in hyperreactive platelets (P=0.05). Among our cohort of 288 subjects, a VAMP8 single-nucleotide polymorphism (rs1010) was associated with platelet reactivity in an age-dependent manner (P<0.003). MicroRNA-96 was predicted to bind to the 3'-untranslated regionof VAMP8 mRNA and was detected in platelets. Overexpression of microRNA-96 in VAMP8-expressing cell lines caused a dose-dependent decrease in VAMP8 protein and mRNA, suggesting a role in VAMP8 mRNA degradation. CONCLUSIONS These findings support a role for VAMP8/endobrevin in the heterogeneity of platelet reactivity, and suggest a role for microRNA-96 in the regulation of VAMP8 expression.
Collapse
Affiliation(s)
- A A Kondkar
- Thomas Jefferson University, The Cardeza Foundation for Hematologic Research and the Department of Medicine, Jefferson Medical College, Philadelphia, PA 19107, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
134
|
Schubert P, Devine DV. Proteomics meets blood banking: identification of protein targets for the improvement of platelet quality. J Proteomics 2010; 73:436-44. [PMID: 19683081 DOI: 10.1016/j.jprot.2009.08.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2009] [Revised: 07/11/2009] [Accepted: 08/04/2009] [Indexed: 12/27/2022]
Abstract
Proteomics has brought new perspectives to the fields of hematology and transfusion medicine in the last decade. The steady improvement of proteomic technology is propelling novel discoveries of molecular mechanisms by studying protein expression, post-translational modifications and protein interactions. This review article focuses on the application of proteomics to the identification of molecular mechanisms leading to the deterioration of blood platelets during storage - a critical aspect in the provision of platelet transfusion products. Several proteomic approaches have been employed to analyse changes in the platelet protein profile during storage and the obtained data now need to be translated into platelet biochemistry in order to connect the results to platelet function. Targeted biochemical applications then allow the identification of points for intervention in signal transduction pathways. Once validated and placed in a transfusion context, these data will provide further understanding of the underlying molecular mechanisms leading to platelet storage lesion. Future aspects of proteomics in blood banking will aim to make use of protein markers identified for platelet storage lesion development to monitor proteome changes when alterations such as the use of additive solutions or pathogen reduction strategies are put in place in order to improve platelet quality for patients.
Collapse
Affiliation(s)
- Peter Schubert
- Canadian Blood Services, Centre for Blood Research and the Department of Pathology and Laboratory Medicine, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, Canada
| | | |
Collapse
|
135
|
Bugert P, Klüter H. Das thrombozytäre Transkriptom. Hamostaseologie 2010. [DOI: 10.1007/978-3-642-01544-1_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
136
|
Schick BP. Serglycin proteoglycan deletion in mouse platelets: physiological effects and their implications for platelet contributions to thrombosis, inflammation, atherosclerosis, and metastasis. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2010; 93:235-87. [PMID: 20807648 DOI: 10.1016/s1877-1173(10)93011-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Serglycin is found in all nucleated hematopoietic cells and platelets, blood vessels, various reproductive and developmental tissues, and in chondrocytes. The serglycin knockout mouse has demonstrated that this proteoglycan is required for proper generation and function of secretory granules in several hematopoietic cells. The effects on platelets are profound, and include diminishing platelet aggregation responses and formation of platelet thrombi. This chapter will review cell-specific aspects of serglycin structure, its gene regulation, cell and tissue localization, and the effects of serglycin deletion on hematopoietic cell granule structure and function. The effects of serglycin knockout on platelets are described and discussed in detail. Rationales for further investigations into the contribution of serglycin to the known roles of platelets in thrombosis, inflammation, atherosclerosis, and tumor metastasis are presented.
Collapse
Affiliation(s)
- Barbara P Schick
- Department of Medicine, Cardeza Foundation for Hematologic Research, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
137
|
Abstract
Exploiting the potential of omics for clinical diagnosis, prognosis, and therapeutic purposes has currently been receiving a lot of attention. In recent years, most of the effort has been put into demonstrating the possible clinical applications of the various omics fields. The cost-effectiveness analysis has been, so far, rather neglected. The cost of omics-derived applications is still very high, but future technological improvements are likely to overcome this problem. In this chapter, we will give a general background of the main omics fields and try to provide some examples of the most successful applications of omics that might be used in clinical diagnosis and in a therapeutic context.
Collapse
Affiliation(s)
- Ewa Gubb
- Bioinformatics, Parque Technológico de Bizkaia, Derio, Spain
| | | |
Collapse
|
138
|
Abstract
Criteria for distinguishing among etiologies of thrombocytosis are limited in their capacity to delineate clonal (essential thrombocythemia [ET]) from nonclonal (reactive thrombocytosis [RT]) etiologies. We studied platelet transcript profiles of 126 subjects (48 controls, 38 RT, 40 ET [24 contained the JAK2V(617)F mutation]) to identify transcript subsets that segregated phenotypes. Cross-platform consistency was validated using quantitative real-time polymerase chain reaction (RT-PCR). Class prediction algorithms were developed to assign phenotypic class between the thrombocytosis cohorts, and by JAK2 genotype. Sex differences were rare in normal and ET cohorts (< 1% of genes) but were male-skewed for approximately 3% of RT genes. An 11-biomarker gene subset using the microarray data discriminated among the 3 cohorts with 86.3% accuracy, with 93.6% accuracy in 2-way class prediction (ET vs RT). Subsequent quantitative RT-PCR analysis established that these biomarkers were 87.1% accurate in prospective classification of a new cohort. A 4-biomarker gene subset predicted JAK2 wild-type ET in more than 85% patient samples using either microarray or RT-PCR profiling, with lower predictive capacity in JAK2V(617)F mutant ET patients. These results establish that distinct genetic biomarker subsets can predict thrombocytosis class using routine phlebotomy.
Collapse
|
139
|
Abstract
Although mammalian platelets are anucleated cells, a number of studies have shown that they retain a pool of messenger RNA (mRNA) carried over from the megakaryocyte during thrombopoiesis. Platelet mRNA was originally thought to be relatively unstable and short-lived within the youngest cells and has been used as a potential marker of platelet turnover. In this article we will discuss both theoretical and methodological issues related to the measurement of these younger, "reticulated platelets". A key question relating to platelet mRNA is also whether it has any functional relevance other than a marker of platelet immaturity. Evidence going back more than 30 years suggests that platelets can biosynthesize proteins. However, it is only very recently that the nature and specificity of platelet mRNA has been examined in any detail. Difficulties in obtaining pure platelet mRNA, free of contamination from other cells has added to the complexity of unravelling this story. However, there is now clear evidence that platelets contain small but significant levels of message for a variety of proteins. The platelet mRNA pool is much richer and more diverse than previously thought and recent data suggests that regulated synthesis of a selected number of proteins can be induced on platelet activation. The full complexity of the platelet genome is now just being revealed and may open the possibility for improved diagnosis and therapy of many haemostatic and thrombotic disorders.
Collapse
Affiliation(s)
- Paul Harrison
- Oxford Haemophilia and Thrombosis Centre, Churchill Hospital, Oxford, UK.
| | | |
Collapse
|
140
|
Abstract
The use of molecular diagnostic techniques in clinical and research hemostasis laboratories is increasing as genetic factors that affect the procoagulant and anticoagulant systems are identified. Many of these molecular alterations are associated with thrombotic tendencies, whereas others tip the hemostatic balance in favor of bleeding. In either scenario, molecular testing may serve as a primary diagnostic modality or may provide information that complements clot-based "functional" assays. The clinical application of DNA-based testing continues to expand since the discoveries of the factor V Leiden and prothrombin G20210A gene mutations. Indications for genetic testing continue to evolve as the underlying causes of hemostatic disorders are better understood. Further development of molecular assays depends on their proved utility in the clinical management and treatment of these complex multifactorial disorders.
Collapse
|
141
|
Landry P, Plante I, Ouellet DL, Perron MP, Rousseau G, Provost P. Existence of a microRNA pathway in anucleate platelets. Nat Struct Mol Biol 2009; 16:961-6. [PMID: 19668211 DOI: 10.1038/nsmb.1651] [Citation(s) in RCA: 384] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2009] [Accepted: 07/02/2009] [Indexed: 12/15/2022]
Abstract
Platelets have a crucial role in the maintenance of hemostasis as well as in thrombosis and vessel occlusion, which underlie stroke and acute coronary syndromes. Anucleate platelets contain mRNAs and are capable of protein synthesis, raising the issue of how these mRNAs are regulated. Here we show that human platelets harbor an abundant and diverse array of microRNAs (miRNAs), which are known as key regulators of mRNA translation in other cell types. Further analyses revealed that platelets contain the Dicer and Argonaute 2 (Ago2) complexes, which function in the processing of exogenous miRNA precursors and the control of specific reporter transcripts, respectively. Detection of the receptor P2Y(12) mRNA in Ago2 immunoprecipitates suggests that P2Y(12) expression may be subjected to miRNA control in human platelets. Our study lends an additional level of complexity to the control of gene expression in these anucleate elements of the cardiovascular system.
Collapse
Affiliation(s)
- Patricia Landry
- Centre de Recherche en Rhumatologie et Immunologie, CHUL Research Center/CHUQ, and Faculty of Medicine, Université Laval, Quebec, Quebec, Canada
| | | | | | | | | | | |
Collapse
|
142
|
Possibility of a rebound phenomenon following antiplatelet therapy withdrawal: A look at the clinical and pharmacological evidence. Pharmacol Ther 2009; 123:178-86. [DOI: 10.1016/j.pharmthera.2009.03.019] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2009] [Accepted: 03/20/2009] [Indexed: 02/02/2023]
|
143
|
Kleiman NS, Freedman JE, Tracy PB, Furie BC, Bray PF, Rao SV, Phillips DR, Storey RF, Rusconi CP, French PA, Steinhubl SR, Becker RC. Platelets: Developmental biology, physiology, and translatable platforms for preclinical investigation and drug development. Platelets 2009; 19:239-51. [DOI: 10.1080/09537100801947442] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
144
|
Abstract
Tissue factor (TF) is the most important initiator of intravascular coagulation. This article will review published evidence on the contribution of platelets to TF exposure to the circulating blood. The following mechanisms will be discussed: decryption of monocyte TF by platelets, contribution of platelets to TF expression in leukocytes, platelet-derived TF and its procoagulant activity, and activation of circulating TF by platelets.
Collapse
Affiliation(s)
- W Lösche
- University Hospital Jena, Department of Anaesthesiology and Intensive Care Medicine, Research Centre Labeda, Jena, Germany.
| |
Collapse
|
145
|
Blood-related proteomics. J Proteomics 2009; 73:483-507. [PMID: 19567275 DOI: 10.1016/j.jprot.2009.06.010] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2009] [Revised: 06/17/2009] [Accepted: 06/20/2009] [Indexed: 12/12/2022]
Abstract
Blood-related proteomics is an emerging field, recently gaining momentum. Indeed, a wealth of data is now available and a plethora of groups has contributed to add pieces to the jigsaw puzzle of protein complexity within plasma and blood cells. In this review article we purported to sail across the mare magnum of the actual knowledge in this research endeavour. The main strides in proteomic investigations on red blood cells, platelets, plasma and white blood cells are hereby presented in a chronological order. Moreover, a glance is given at prospective studies which promise to shift the focus of attention from the end product to its provider, the donor, in a sort of Kantian "Copernican revolution". A well-rounded portrait of the usefulness of proteomics in blood-related research is accurately given. In particular, proteomic tools could be adopted to follow the main steps of the blood-banking production processes (a comparison of collection methods, pathogen inactivation techniques, storage protocols). Thus proteomics has been recently transformed from a mere basic-research extremely-expensive toy into a dramatically-sensitive and efficient eye-lens to either delve into the depths of the molecular mechanisms of blood and blood components or to establish quality parameters in the blood-banking production chain totally anew.
Collapse
|
146
|
Schubert P, Thon JN, Walsh GM, Chen CHI, Moore ED, Devine DV, Kast J. A signaling pathway contributing to platelet storage lesion development: targeting PI3-kinase-dependent Rap1 activation slows storage-induced platelet deterioration. Transfusion 2009; 49:1944-55. [PMID: 19497060 DOI: 10.1111/j.1537-2995.2009.02224.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND The term platelet storage lesion (PSL) describes the structural and biochemical changes in platelets (PLTs) during storage. These are typified by alterations of morphologic features and PLT metabolism leading to reduced functionality and hence reduced viability for transfusion. While the manifestations of the storage lesion are well characterized, the biochemical pathways involved in the initiation of this process are unknown. STUDY DESIGN AND METHODS A complementary proteomic approach has recently been applied to analyze changes in the PLT proteome during storage. By employing stringent proteomic criteria, 12 proteins were identified as significantly and consistently changing in relative concentration over a 7-day storage period. Microscopy, Western blot analysis, flow cytometry, and PLT functionality analyses were used to unravel the involvement of a subset of these 12 proteins, which are connected through integrin signaling in one potential signaling pathway underlying storage lesion development. RESULTS Microscopic analysis revealed changes in localization of glycoprotein IIIa, Rap1, and talin during storage. Rap1 activation was observed to correlate with expression of the PLT activation marker CD62P. PLTs incubated for 7 days with the PI3-kinase inhibitor LY294002 showed diminished Rap1 activation as well as a moderate reduction in integrin alphaIIbbeta3 activation and release of alpha-granules. Furthermore, this inhibitor seemed to improve PLT integrity and quality during storage as several in vitro probes showed a deceleration of PLT activation. CONCLUSION These results provide the first evidence for a signaling pathway mediating PSL in which PI3-kinase-dependent Rap1 activation leads to integrin alphaIIbbeta3 activation and PLT degranulation.
Collapse
Affiliation(s)
- Peter Schubert
- Canadian Blood Services, University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | | | | | | | | |
Collapse
|
147
|
Power KA, McRedmond JP, de Stefani A, Gallagher WM, Ó Gaora P. High-throughput proteomics detection of novel splice isoforms in human platelets. PLoS One 2009; 4:e5001. [PMID: 19308253 PMCID: PMC2654914 DOI: 10.1371/journal.pone.0005001] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2008] [Accepted: 02/20/2009] [Indexed: 12/16/2022] Open
Abstract
Alternative splicing (AS) is an intrinsic regulatory mechanism of all metazoans. Recent findings suggest that 100% of multiexonic human genes give rise to splice isoforms. AS can be specific to tissue type, environment or developmentally regulated. Splice variants have also been implicated in various diseases including cancer. Detection of these variants will enhance our understanding of the complexity of the human genome and provide disease-specific and prognostic biomarkers. We adopted a proteomics approach to identify exon skip events - the most common form of AS. We constructed a database harboring the peptide sequences derived from all hypothetical exon skip junctions in the human genome. Searching tandem mass spectrometry (MS/MS) data against the database allows the detection of exon skip events, directly at the protein level. Here we describe the application of this approach to human platelets, including the mRNA-based verification of novel splice isoforms of ITGA2, NPEPPS and FH. This methodology is applicable to all new or existing MS/MS datasets.
Collapse
Affiliation(s)
- Karen A. Power
- UCD Conway Institute and UCD School of Biomolecular & Biomedical Sciences, UCD Conway Institute, University College Dublin, Belfield, Dublin, Ireland
| | - James P. McRedmond
- UCD Conway Institute and UCD School of Biomolecular & Biomedical Sciences, UCD Conway Institute, University College Dublin, Belfield, Dublin, Ireland
| | | | - William M. Gallagher
- UCD Conway Institute and UCD School of Biomolecular & Biomedical Sciences, UCD Conway Institute, University College Dublin, Belfield, Dublin, Ireland
| | - Peadar Ó Gaora
- UCD Conway Institute and UCD School of Medicine & Medical Sciences, UCD Conway Institute, University College Dublin, Belfield, Dublin, Ireland
- * E-mail:
| |
Collapse
|
148
|
Weyrich AS, Schwertz H, Kraiss LW, Zimmerman GA. Protein synthesis by platelets: historical and new perspectives. J Thromb Haemost 2009; 7:241-6. [PMID: 18983498 PMCID: PMC3027201 DOI: 10.1111/j.1538-7836.2008.03211.x] [Citation(s) in RCA: 207] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
In the late 1960s, numerous investigators independently demonstrated that platelets are capable of synthesizing proteins. Studies continued at a steady pace over the next 30 years and into the 21st century. Collectively, these investigations confirmed that platelets synthesize proteins and that the pattern of protein synthesis changes in response to cellular activation. More recent studies have characterized the mechanisms by which platelets synthesize proteins and have shown that protein synthesis alters the phenotype and functions of platelets. Here, we chronologically review our increased understanding of protein synthetic responses in platelets and discuss how the field may evolve over the next decade.
Collapse
Affiliation(s)
- A S Weyrich
- Department of Internal Medicine, University of Utah, Salt Lake City, UT 84112, USA.
| | | | | | | |
Collapse
|
149
|
Greening DW, Glenister KM, Sparrow RL, Simpson RJ. Enrichment of human platelet membranes for proteomic analysis. Methods Mol Biol 2009; 528:245-258. [PMID: 19153697 DOI: 10.1007/978-1-60327-310-7_17] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Platelets (thrombocytes) are the smallest human blood cells and are pivotal in processes of hemostasis and thrombosis. Central to their function, the activation of platelets includes a complex interplay of adhesion and signalling molecules mediated via the plasma and inner membrane. Because platelets are enucleated, the analysis of the proteome is the best way to approach their biology. Here, we employ mass spectrometry (MS)-based proteomics to characterise membrane proteins derived from non-stimulated human platelets. This protocol details the extraction and purification of platelet membrane proteins from whole blood using SDS-PAGE in conjunction with LC-MS/MS. This method allowed the identification, and characterization of 207 platelet membrane proteins (PMP) from approximately 9.95 x 10(9) platelets (16).
Collapse
|
150
|
Transcript profiling of human platelets using microarray and serial analysis of gene expression (SAGE). Methods Mol Biol 2009; 496:245-72. [PMID: 18839115 DOI: 10.1007/978-1-59745-553-4_16] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Platelets are anucleated cells that are generated from megakaryocytes via thrombopoiesis. They lack genomic DNA but have a pool of individual mRNA transcripts. Taken together, these mRNAs constitute a platelet transcriptome. Platelets have a unique and reproducible transcript profile, which includes approximately 1,600-3,000 individual transcripts. In this chapter, we will focus on platelet purification and on transcript profiling using an Affymetrix microarray platform and serial analysis of gene expression (SAGE). Platelet purification is described in detail. Large-scale platelet purification schema is designed to purify platelets from apheresis platelet bags (approximately 3-5 x 10(11) platelets/bag). Modification of this schema --small-scale platelet purification--is designed to isolate platelets from 20 ml of peripheral blood. This chapter provides detailed protocols for microarray and SAGE transcript profiling. We also discuss peculiarities of platelet purification, RNA isolation, and transcript profiling.
Collapse
|