51
|
Unsworth AJ, Bye AP, Sage T, Gaspar RS, Eaton N, Drew C, Stainer A, Kriek N, Volberding PJ, Hutchinson JL, Riley R, Jones S, Mundell SJ, Cui W, Falet H, Gibbins JM. Antiplatelet properties of Pim kinase inhibition are mediated through disruption of thromboxane A2 receptor signaling. Haematologica 2021; 106:1968-1978. [PMID: 32467143 PMCID: PMC8252961 DOI: 10.3324/haematol.2019.223529] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Indexed: 12/17/2022] Open
Abstract
Pim kinases are upregulated in several forms of cancer, contributing to cell survival and tumor development, but their role in platelet function and thrombotic disease has not been explored. We report for the first time that Pim-1 kinase is expressed in human and mouse platelets. Genetic deletion or pharmacological inhibition of Pim kinase results in reduced thrombus formation but is not associated with impaired hemostasis. Attenuation of thrombus formation was found to be due to inhibition of the thromboxane A2 receptor as effects on platelet function were non-additive to inhibition caused by the cyclo-oxygenase inhibitor indomethacin or the thromboxane A2 receptor antagonist GR32191. Treatment with Pim kinase inhibitors caused reduced surface expression of the thromboxane A2 receptor and resulted in reduced responses to thromboxane A2 receptor agonists, indicating a role for Pim kinase in the regulation of thromboxane A2 receptor function. Our research identifies a novel, Pim kinase-dependent regulatory mechanism for the thromboxane A2 receptor and represents a new targeting strategy that is independent of cyclo-oxygenase-1 inhibition or direct antagonism of the thromboxane A2 receptor that, while attenuating thrombosis, does not increase bleeding.
Collapse
Affiliation(s)
- Amanda J Unsworth
- University of Reading and Dept. of Life Sciences, Manchester Metropolitan University Manchester, UK
| | - Alexander P Bye
- Institute for Cardiovascular, Metabolic Research, University of Reading, Reading, UK
| | - Tanya Sage
- Institute for Cardiovascular and Metabolic Research, University of Reading, Reading, UK
| | - Renato S Gaspar
- Institute for Cardiovascular and Metabolic Research, University of Reading, Reading, UK
| | - Nathan Eaton
- Blood Research Institute and Medical College of Wisconsin, Versiti, Milwaukee, WI, USA
| | - Caleb Drew
- Blood Research Institute, Versiti, Milwaukee, WI, USA
| | - Alexander Stainer
- Institute for Cardiovascular and Metabolic Research, University of Reading, Reading, UK
| | - Neline Kriek
- Institute for Cardiovascular and Metabolic Research, University of Reading, Reading, UK
| | - Peter J Volberding
- Blood Research Institute and Medical College of Wisconsin, Versiti, Milwaukee, WI, USA
| | - James L Hutchinson
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Ryan Riley
- Department of Life Sciences, Manchester Metropolitan University, Manchester, UK
| | - Sarah Jones
- Department of Life Sciences, Manchester Metropolitan University, Manchester, UK
| | - Stuart J Mundell
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Weiguo Cui
- Blood Research Institute, Versiti and Medical College of Wisconsin, Milwaukee, WI, USA
| | - Hervé Falet
- Blood Research Institute, Versiti and Medical College of Wisconsin, Milwaukee, WI, USA
| | - Jonathan M Gibbins
- Institute for Cardiovascular and Metabolic Research, University of Reading, Reading, UK
| |
Collapse
|
52
|
Ramos‐Marquès E, García‐Mendívil L, Pérez‐Zabalza M, Santander‐Badules H, Srinivasan S, Oliveros JC, Torres‐Pérez R, Cebollada A, Vallejo‐Gil JM, Fresneda‐Roldán PC, Fañanás‐Mastral J, Vázquez‐Sancho M, Matamala‐Adell M, Sorribas‐Berjón JF, Bellido‑Morales JA, Mancebón‑Sierra FJ, Vaca‑Núñez AS, Ballester‐Cuenca C, Jiménez‐Navarro M, Villaescusa JM, Garrido‐Huéscar E, Segovia‐Roldán M, Oliván‐Viguera A, Gómez‐González C, Muñiz G, Diez E, Ordovás L, Pueyo E. Chronological and biological aging of the human left ventricular myocardium: Analysis of microRNAs contribution. Aging Cell 2021; 20:e13383. [PMID: 34092006 PMCID: PMC8282276 DOI: 10.1111/acel.13383] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/26/2021] [Accepted: 04/26/2021] [Indexed: 11/29/2022] Open
Abstract
Aging is the main risk factor for cardiovascular diseases. In humans, cardiac aging remains poorly characterized. Most studies are based on chronological age (CA) and disregard biological age (BA), the actual physiological age (result of the aging rate on the organ structure and function), thus yielding potentially imperfect outcomes. Deciphering the molecular basis of ventricular aging, especially by BA, could lead to major progresses in cardiac research. We aim to describe the transcriptome dynamics of the aging left ventricle (LV) in humans according to both CA and BA and characterize the contribution of microRNAs, key transcriptional regulators. BA is measured using two CA-associated transcriptional markers: CDKN2A expression, a cell senescence marker, and apparent age (AppAge), a highly complex transcriptional index. Bioinformatics analysis of 132 LV samples shows that CDKN2A expression and AppAge represent transcriptomic changes better than CA. Both BA markers are biologically validated in relation to an aging phenotype associated with heart dysfunction, the amount of cardiac fibrosis. BA-based analyses uncover depleted cardiac-specific processes, among other relevant functions, that are undetected by CA. Twenty BA-related microRNAs are identified, and two of them highly heart-enriched that are present in plasma. We describe a microRNA-gene regulatory network related to cardiac processes that are partially validated in vitro and in LV samples from living donors. We prove the higher sensitivity of BA over CA to explain transcriptomic changes in the aging myocardium and report novel molecular insights into human LV biological aging. Our results can find application in future therapeutic and biomarker research.
Collapse
Affiliation(s)
- Estel Ramos‐Marquès
- Biomedical Signal Interpretation and Computational Simulation group (BSICoS) Aragón Institute of Engineering Research University of Zaragoza Zaragoza Spain
- BSICoSIIS Aragón Zaragoza Spain
| | - Laura García‐Mendívil
- Biomedical Signal Interpretation and Computational Simulation group (BSICoS) Aragón Institute of Engineering Research University of Zaragoza Zaragoza Spain
- BSICoSIIS Aragón Zaragoza Spain
| | - María Pérez‐Zabalza
- Biomedical Signal Interpretation and Computational Simulation group (BSICoS) Aragón Institute of Engineering Research University of Zaragoza Zaragoza Spain
- BSICoSIIS Aragón Zaragoza Spain
| | - Hazel Santander‐Badules
- Biomedical Signal Interpretation and Computational Simulation group (BSICoS) Aragón Institute of Engineering Research University of Zaragoza Zaragoza Spain
| | - Sabarathinam Srinivasan
- Biomedical Signal Interpretation and Computational Simulation group (BSICoS) Aragón Institute of Engineering Research University of Zaragoza Zaragoza Spain
- BSICoSIIS Aragón Zaragoza Spain
| | - Juan Carlos Oliveros
- Bioinformatics for Genomics and Proteomics National Center of Biotechnology‐ Spanish National Research Council Madrid Spain
| | - Rafael Torres‐Pérez
- Bioinformatics for Genomics and Proteomics National Center of Biotechnology‐ Spanish National Research Council Madrid Spain
| | | | | | | | | | - Manuel Vázquez‐Sancho
- Department of Cardiovascular Surgery University Hospital Miguel Servet Zaragoza Spain
| | - Marta Matamala‐Adell
- Department of Cardiovascular Surgery University Hospital Miguel Servet Zaragoza Spain
| | | | | | | | | | | | - Manuel Jiménez‐Navarro
- Heart Area Hospital Clínico Universitario Virgen de la Victoria, CIBERCV IBIMA, Universidad de Málaga, UMA Málaga Spain
| | - José Manuel Villaescusa
- UGC Heart Area Cardiovascular Surgery Department Hospital Universitario Virgen de la Victoria de Málaga Fundación Pública Andaluza para la Investigación de Málaga en Biomedicina y Salud (FIMABIS) CIBERCV Enfermedades Cardiovasculares Instituto de Salud Carlos III University of Málaga Madrid Spain
| | - Elisa Garrido‐Huéscar
- Biomedical Signal Interpretation and Computational Simulation group (BSICoS) Aragón Institute of Engineering Research University of Zaragoza Zaragoza Spain
- BSICoSIIS Aragón Zaragoza Spain
| | - Margarita Segovia‐Roldán
- Biomedical Signal Interpretation and Computational Simulation group (BSICoS) Aragón Institute of Engineering Research University of Zaragoza Zaragoza Spain
- BSICoSIIS Aragón Zaragoza Spain
| | - Aida Oliván‐Viguera
- Biomedical Signal Interpretation and Computational Simulation group (BSICoS) Aragón Institute of Engineering Research University of Zaragoza Zaragoza Spain
- BSICoSIIS Aragón Zaragoza Spain
| | | | - Gorka Muñiz
- Department of Pathology San Jorge Hospital Huesca Spain
| | - Emiliano Diez
- Institute of Experimental Medicine and Biology of Cuyo (IMBECU) CONICET Mendoza Argentina
| | - Laura Ordovás
- Biomedical Signal Interpretation and Computational Simulation group (BSICoS) Aragón Institute of Engineering Research University of Zaragoza Zaragoza Spain
- BSICoSIIS Aragón Zaragoza Spain
- ARAID Foundation Zaragoza Spain
| | - Esther Pueyo
- Biomedical Signal Interpretation and Computational Simulation group (BSICoS) Aragón Institute of Engineering Research University of Zaragoza Zaragoza Spain
- BSICoSIIS Aragón Zaragoza Spain
- Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine (CIBER‐BBN) Zaragoza Spain
| |
Collapse
|
53
|
Garcia A, Dunoyer-Geindre S, Fontana P. Do miRNAs Have a Role in Platelet Function Regulation? Hamostaseologie 2021; 41:217-224. [PMID: 34192780 DOI: 10.1055/a-1478-2105] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
MicroRNAs (miRNAs) are a class of non-coding RNAs known to repress mRNA translation and subsequent protein production. miRNAs are predicted to modulate many targets and are involved in regulating various cellular processes. Identifying their role in cell function regulation may allow circulating miRNAs to be used as diagnostic or prognostic markers of various diseases. Increasing numbers of clinical studies have shown associations between circulating miRNA levels and platelet reactivity or the recurrence of cardiovascular events. However, these studies differed regarding population selection, sample types used, miRNA quantification procedures, and platelet function assays. Furthermore, they often lacked functional validation of the miRNA identified in such studies. The latter step is essential to identifying causal relationships and understanding if and how miRNAs regulate platelet function. This review describes recent advances in translational research dedicated to identifying miRNAs' roles in platelet function regulation.
Collapse
Affiliation(s)
- A Garcia
- Geneva Platelet Group, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | | | - P Fontana
- Geneva Platelet Group, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Division of Angiology and Haemostasis, Geneva University Hospitals, Geneva, Switzerland
| |
Collapse
|
54
|
Ali H, Harting R, de Vries R, Ali M, Wurdinger T, Best MG. Blood-Based Biomarkers for Glioma in the Context of Gliomagenesis: A Systematic Review. Front Oncol 2021; 11:665235. [PMID: 34150629 PMCID: PMC8211985 DOI: 10.3389/fonc.2021.665235] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 05/18/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Gliomas are the most common and aggressive tumors of the central nervous system. A robust and widely used blood-based biomarker for glioma has not yet been identified. In recent years, a plethora of new research on blood-based biomarkers for glial tumors has been published. In this review, we question which molecules, including proteins, nucleic acids, circulating cells, and metabolomics, are most promising blood-based biomarkers for glioma diagnosis, prognosis, monitoring and other purposes, and align them to the seminal processes of cancer. METHODS The Pubmed and Embase databases were systematically searched. Biomarkers were categorized in the identified biomolecules and biosources. Biomarker characteristics were assessed using the area under the curve (AUC), accuracy, sensitivity and/or specificity values and the degree of statistical significance among the assessed clinical groups was reported. RESULTS 7,919 references were identified: 3,596 in PubMed and 4,323 in Embase. Following screening of titles, abstracts and availability of full-text, 262 articles were included in the final systematic review. Panels of multiple biomarkers together consistently reached AUCs >0.8 and accuracies >80% for various purposes but especially for diagnostics. The accuracy of single biomarkers, consisting of only one measurement, was far more variable, but single microRNAs and proteins are generally more promising as compared to other biomarker types. CONCLUSION Panels of microRNAs and proteins are most promising biomarkers, while single biomarkers such as GFAP, IL-10 and individual miRNAs also hold promise. It is possible that panels are more accurate once these are involved in different, complementary cancer-related molecular pathways, because not all pathways may be dysregulated in cancer patients. As biomarkers seem to be increasingly dysregulated in patients with short survival, higher tumor grades and more pathological tumor types, it can be hypothesized that more pathways are dysregulated as the degree of malignancy of the glial tumor increases. Despite, none of the biomarkers found in the literature search seem to be currently ready for clinical implementation, and most of the studies report only preliminary application of the identified biomarkers. Hence, large-scale validation of currently identified and potential novel biomarkers to show clinical utility is warranted.
Collapse
Affiliation(s)
- Hamza Ali
- Department of Neurosurgery, Brain Tumor Center Amsterdam, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center and Academic Medical Center, Amsterdam, Netherlands
| | - Romée Harting
- Department of Neurosurgery, Brain Tumor Center Amsterdam, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center and Academic Medical Center, Amsterdam, Netherlands
| | - Ralph de Vries
- Medical Library, Vrije Universiteit, Amsterdam, Netherlands
| | - Meedie Ali
- Department of Neurosurgery, Brain Tumor Center Amsterdam, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center and Academic Medical Center, Amsterdam, Netherlands
| | - Thomas Wurdinger
- Department of Neurosurgery, Brain Tumor Center Amsterdam, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center and Academic Medical Center, Amsterdam, Netherlands
| | - Myron G. Best
- Department of Neurosurgery, Brain Tumor Center Amsterdam, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center and Academic Medical Center, Amsterdam, Netherlands
| |
Collapse
|
55
|
Platelets as drivers of ischemia/reperfusion injury after stroke. Blood Adv 2021; 5:1576-1584. [PMID: 33687431 DOI: 10.1182/bloodadvances.2020002888] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 02/01/2021] [Indexed: 12/14/2022] Open
Abstract
Ischemic stroke is a leading cause of morbidity and mortality worldwide and, despite reperfusion either via thrombolysis or thrombectomy, stroke patients often suffer from lifelong disabilities. These persistent neurological deficits may be improved by treating the ischemia/reperfusion (I/R) injury that occurs following ischemic stroke. There are currently no approved therapies to treat I/R injury, and thus it is imperative to find new targets to decrease the burden of ischemic stroke and related diseases. Platelets, cell fragments from megakaryocytes, are primarily known for their role in hemostasis. More recently, investigators have studied the nonhemostatic role of platelets in inflammatory pathologies, such as I/R injury after ischemic stroke. In this review, we seek to provide an overview of how I/R can lead to platelet activation and how activated platelets, in turn, can exacerbate I/R injury after stroke. We will also discuss potential mechanisms by which platelets may ameliorate I/R injury.
Collapse
|
56
|
Platelets and extracellular vesicles and their cross talk with cancer. Blood 2021; 137:3192-3200. [PMID: 33940593 DOI: 10.1182/blood.2019004119] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 11/16/2020] [Indexed: 02/06/2023] Open
Abstract
Platelets play significant and varied roles in cancer progression, as detailed throughout this review series, via direct interactions with cancer cells and by long-range indirect interactions mediated by platelet releasates. Microvesicles (MVs; also referred to as microparticles) released from activated platelets have emerged as major contributors to the platelet-cancer nexus. Interactions of platelet-derived MVs (PMVs) with cancer cells can promote disease progression through multiple mechanisms, but PMVs also harbor antitumor functions. This complex relationship derives from PMVs' binding to both cancer cells and nontransformed cells in the tumor microenvironment and transferring platelet-derived contents to the target cell, each of which can have stimulatory or modulatory effects. MVs are extracellular vesicles of heterogeneous size, ranging from 100 nm to 1 µm in diameter, shed by living cells during the outward budding of the plasma membrane, entrapping local cytosolic contents in an apparently stochastic manner. Hence, PMVs are encapsulated by a lipid bilayer harboring surface proteins and lipids mirroring the platelet exterior, with internal components including platelet-derived mature messenger RNAs, pre-mRNAs, microRNAs, and other noncoding RNAs, proteins, second messengers, and mitochondria. Each of these elements engages in established and putative PMV functions in cancer. In addition, PMVs contribute to cancer comorbidities because of their roles in coagulation and thrombosis and via interactions with inflammatory cells. However, separating the effects of PMVs from those of platelets in cancer contexts continues to be a major hurdle. This review summarizes our emerging understanding of the complex roles of PMVs in the development and progression of cancer and cancer comorbidities.
Collapse
|
57
|
Garcia A, Dunoyer-Geindre S, Nolli S, Reny JL, Fontana P. An Ex Vivo and In Silico Study Providing Insights into the Interplay of Circulating miRNAs Level, Platelet Reactivity and Thrombin Generation: Looking beyond Traditional Pharmacogenetics. J Pers Med 2021; 11:jpm11050323. [PMID: 33919053 PMCID: PMC8143175 DOI: 10.3390/jpm11050323] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 04/14/2021] [Accepted: 04/19/2021] [Indexed: 12/18/2022] Open
Abstract
Platelet reactivity (PR), a key pharmacodynamic (PD) component of the action of antiplatelet drugs in cardiovascular disease (CVD) patients, is highly variable. PR is associated with occurrence or recurrence of thrombotic and bleeding events, but this association is modulated by several factors. Conventional pharmacogenetics explains a minor part of this PR variability, and among determinants of PR, circulating microRNAs (miRNAs) have been the focus of attention during these last years as biomarkers to predict PR and clinical outcomes in CVD. This being said, the impact of miRNAs on platelet function and the mechanisms behind it are largely unknown. The level of a set of candidate miRNAs including miR-126-3p, miR-150-5p, miR-204-5p and miR-223-3p was quantified in plasma samples of stable CVD patients and correlated with PR as assessed by light-transmission aggregometry and in vivo thrombin generation markers. Finally, miRNA target networks were built based on genes involved in platelet function. We show that all candidate miRNAs were associated with platelet aggregation, while only miR-126-3p and miR-223-3p were positively correlated with in vivo thrombin generation markers. In silico analysis identified putative miRNA targets involved in platelet function regulation. Circulating miRNAs were associated with different aspects of platelet reactivity, including platelet aggregation and platelet-supported thrombin generation. This paves the way to a personalized antithrombotic treatment according to miRNA profile in CVD patients.
Collapse
Affiliation(s)
- Alix Garcia
- Geneva Platelet Group, Faculty of Medicine, University of Geneva, 1205 Geneva, Switzerland; (A.G.); (S.D.-G.); (S.N.); (J.-L.R.)
| | - Sylvie Dunoyer-Geindre
- Geneva Platelet Group, Faculty of Medicine, University of Geneva, 1205 Geneva, Switzerland; (A.G.); (S.D.-G.); (S.N.); (J.-L.R.)
| | - Séverine Nolli
- Geneva Platelet Group, Faculty of Medicine, University of Geneva, 1205 Geneva, Switzerland; (A.G.); (S.D.-G.); (S.N.); (J.-L.R.)
| | - Jean-Luc Reny
- Geneva Platelet Group, Faculty of Medicine, University of Geneva, 1205 Geneva, Switzerland; (A.G.); (S.D.-G.); (S.N.); (J.-L.R.)
- Division of General Internal Medicine, Geneva University Hospitals, 1205 Geneva, Switzerland
| | - Pierre Fontana
- Geneva Platelet Group, Faculty of Medicine, University of Geneva, 1205 Geneva, Switzerland; (A.G.); (S.D.-G.); (S.N.); (J.-L.R.)
- Division of Angiology and Haemostasis, Geneva University Hospitals, 1205 Geneva, Switzerland
- Correspondence: ; Tel.: +41-22-372-97-51
| |
Collapse
|
58
|
Tsamou M, Vrijens K, Wang C, Winckelmans E, Neven KY, Madhloum N, de Kok TM, Nawrot TS. Genome-wide microRNA expression analysis in human placenta reveals sex-specific patterns: an ENVIR ONAGE birth cohort study. Epigenetics 2021; 16:373-388. [PMID: 32892695 PMCID: PMC7993149 DOI: 10.1080/15592294.2020.1803467] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 07/03/2020] [Accepted: 07/24/2020] [Indexed: 12/22/2022] Open
Abstract
There is an increasing interest in microRNAs (miRNAs) as they are of utmost importance in gene regulation at the posttranscriptional level. Sex-related susceptibility for non-communicable diseases later in life could originate in early life. Until now, no data on sex-specific miRNA expression are available for the placenta. Therefore, we investigated the difference by sex of newborn's miRNA expression in human placental tissue. Within the ENVIRONAGE birth cohort, miRNA and mRNA expression profiling was performed in 60 placentae (50% boys) using Agilent (8 × 60 K) microarrays. The distribution of chromosome locations was studied and pathway analysis of the identified sex-specific miRNAs in the placenta was carried out. Of the total 2558 miRNAs on the array, 597 miRNAs were expressed in over 70% of the samples and were included for further analyses. A total of 142 miRNAs were significantly (FDR<0.05) associated with the newborn's sex. In newborn girls, 76 miRNAs had higher expression (hsa-miR-361-5p as most significant) and 66 miRNAs had lower expression (hsa-miR-4646-5p as most significant) than in newborn boys. In the same study population, placental differentially expressed genes by sex were also identified using a whole genome approach. The placental gene expression revealed 27 differentially expressed genes by comparing girls to boys. Ultimately, we studied the miRNA-RNA interactome and identified 14 miRNA-mRNA interactions as sex-specific. Sex differences in placental m(i)RNA expression may reveal sex-specific patterns already present during pregnancy, which may influence physiological conditions in early or later life. These molecular processes might play a role in sex-specific disease susceptibility in later life.
Collapse
Affiliation(s)
- Maria Tsamou
- Center for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - Karen Vrijens
- Center for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - Congrong Wang
- Center for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - Ellen Winckelmans
- Center for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - Kristof Y. Neven
- Center for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - Narjes Madhloum
- Center for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - Theo M. de Kok
- Department of Toxicogenomics, GROW Institute of Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Tim S. Nawrot
- Center for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
- Department of Public Health, Environment & Health Unit, Leuven University (KU Leuven), Leuven, Belgium
| |
Collapse
|
59
|
Garcia A, Dunoyer-Geindre S, Fish RJ, Neerman-Arbez M, Reny JL, Fontana P. Methods to Investigate miRNA Function: Focus on Platelet Reactivity. Thromb Haemost 2021; 121:409-421. [PMID: 33124028 PMCID: PMC8263142 DOI: 10.1055/s-0040-1718730] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 09/08/2020] [Indexed: 12/14/2022]
Abstract
MicroRNAs (miRNAs) are small noncoding RNAs modulating protein production. They are key players in regulation of cell function and are considered as biomarkers in several diseases. The identification of the proteins they regulate, and their impact on cell physiology, may delineate their role as diagnostic or prognostic markers and identify new therapeutic strategies. During the last 3 decades, development of a large panel of techniques has given rise to multiple models dedicated to the study of miRNAs. Since plasma samples are easily accessible, circulating miRNAs can be studied in clinical trials. To quantify miRNAs in numerous plasma samples, the choice of extraction and purification techniques, as well as normalization procedures, are important for comparisons of miRNA levels in populations and over time. Recent advances in bioinformatics provide tools to identify putative miRNAs targets that can then be validated with dedicated assays. In vitro and in vivo approaches aim to functionally validate candidate miRNAs from correlations and to understand their impact on cellular processes. This review describes the advantages and pitfalls of the available techniques for translational research to study miRNAs with a focus on their role in regulating platelet reactivity.
Collapse
Affiliation(s)
- Alix Garcia
- Geneva Platelet Group, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | | | - Richard J. Fish
- Department of Genetic Medicine and Development, University of Geneva, Geneva, Switzerland
| | - Marguerite Neerman-Arbez
- Department of Genetic Medicine and Development, University of Geneva, Geneva, Switzerland
- iGE3, Institute of Genetics and Genomics in Geneva, Geneva, Switzerland
| | - Jean-Luc Reny
- Geneva Platelet Group, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Division of General Internal Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Pierre Fontana
- Geneva Platelet Group, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Division of Angiology and Haemostasis, Geneva University Hospitals, Geneva, Switzerland
| |
Collapse
|
60
|
Pellegrino-Coppola D, Claringbould A, Stutvoet M, Boomsma DI, Ikram MA, Slagboom PE, Westra HJ, Franke L. Correction for both common and rare cell types in blood is important to identify genes that correlate with age. BMC Genomics 2021; 22:184. [PMID: 33722199 PMCID: PMC7958454 DOI: 10.1186/s12864-020-07344-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 12/22/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Aging is a multifactorial process that affects multiple tissues and is characterized by changes in homeostasis over time, leading to increased morbidity. Whole blood gene expression signatures have been associated with aging and have been used to gain information on its biological mechanisms, which are still not fully understood. However, blood is composed of many cell types whose proportions in blood vary with age. As a result, previously observed associations between gene expression levels and aging might be driven by cell type composition rather than intracellular aging mechanisms. To overcome this, previous aging studies already accounted for major cell types, but the possibility that the reported associations are false positives driven by less prevalent cell subtypes remains. RESULTS Here, we compared the regression model from our previous work to an extended model that corrects for 33 additional white blood cell subtypes. Both models were applied to whole blood gene expression data from 3165 individuals belonging to the general population (age range of 18-81 years). We evaluated that the new model is a better fit for the data and it identified fewer genes associated with aging (625, compared to the 2808 of the initial model; P ≤ 2.5⨯10-6). Moreover, 511 genes (~ 18% of the 2808 genes identified by the initial model) were found using both models, indicating that the other previously reported genes could be proxies for less abundant cell types. In particular, functional enrichment of the genes identified by the new model highlighted pathways and GO terms specifically associated with platelet activity. CONCLUSIONS We conclude that gene expression analyses in blood strongly benefit from correction for both common and rare blood cell types, and recommend using blood-cell count estimates as standard covariates when studying whole blood gene expression.
Collapse
Affiliation(s)
| | - Annique Claringbould
- Department of Genetics, University Medical Centre Groningen, Groningen, The Netherlands
| | - Maartje Stutvoet
- Department of Genetics, University Medical Centre Groningen, Groningen, The Netherlands
| | | | - Dorret I Boomsma
- Department of Biological Psychology, Netherlands Twin Register, Amsterdam Public Health research institute and Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - P Eline Slagboom
- Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
| | - Harm-Jan Westra
- Department of Genetics, University Medical Centre Groningen, Groningen, The Netherlands
| | - Lude Franke
- Department of Genetics, University Medical Centre Groningen, Groningen, The Netherlands.
| |
Collapse
|
61
|
Li X, Liu L, Song X, Wang K, Niu L, Xie L, Song X. TEP linc-GTF2H2-1, RP3-466P17.2, and lnc-ST8SIA4-12 as novel biomarkers for lung cancer diagnosis and progression prediction. J Cancer Res Clin Oncol 2021; 147:1609-1622. [PMID: 33792796 DOI: 10.1007/s00432-020-03502-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 12/12/2020] [Indexed: 11/27/2022]
Abstract
PURPOSE Platelets contain a rich repertoire of RNA species, such as mRNAs and long non-coding RNAs. During the development of tumors, platelets are "educated" by cancer cells, altering their transcriptome and molecular content, thereby, tumor educated platelet (TEP) lncRNA profile has the potential to diagnose lung cancer. The current study was aimed to examine whether TEPs might be a potential biomarker for lung cancer. METHODS Platelet precipitation was obtained by low-speed centrifugation. TEP linc-GTF2H2-1, RP3-466P17.2, and lnc-ST8SIA4-12 were selected by lncRNA microarray and validated by qPCR in a large cohort of lung cancer patients and healthy donors. Besides, we analyzed the association of their expression levels with clinicopathological features. RESULTS TEP linc-GTF2H2-1 and RP3-466P17.2 were significantly downregulated, while lnc-ST8SIA4-12 was significantly upregulated in patients with lung cancer or with early-stage lung cancer as compared to healthy donors, possessing AUCs of 0.781, 0.788, 0.725 for lung cancer and 0.704, 0.771, 0.768 for early-stage lung cancer, respectively. Notably, their combination demonstrated the markedly elevated AUCs of 0.921 for lung cancer and 0.895 for early-stage lung cancer. Besides, the combination of TEP linc-GTF2H2-1 was capable to facilitate diagnostic efficiencies of CEA, Cyfra21-1, or NSE to distinguish advanced-stage lung cancer patients from early ones, with an AUC of 0.899 based on the integration of these four factors. CONCLUSION Our data suggested that lncRNAs sequestered in TEPs enabled blood-based lung cancer diagnosis and progression prediction.
Collapse
Affiliation(s)
- Xinyi Li
- Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Ji-Yan Road, Jinan, 250117, Shandong Province, People's Republic of China
| | - Lele Liu
- Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Ji-Yan Road, Jinan, 250117, Shandong Province, People's Republic of China.,Department of Clinical Laboratory, Kunshan Hospital of Traditional Chinese Medicine, Kunshan, Jiangsu, People's Republic of China
| | - Xingguo Song
- Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Ji-Yan Road, Jinan, 250117, Shandong Province, People's Republic of China.,Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, People's Republic of China
| | - Kangyu Wang
- Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Ji-Yan Road, Jinan, 250117, Shandong Province, People's Republic of China
| | - Limin Niu
- Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Ji-Yan Road, Jinan, 250117, Shandong Province, People's Republic of China
| | - Li Xie
- Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Ji-Yan Road, Jinan, 250117, Shandong Province, People's Republic of China
| | - Xianrang Song
- Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Ji-Yan Road, Jinan, 250117, Shandong Province, People's Republic of China. .,Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, People's Republic of China.
| |
Collapse
|
62
|
Anka Idrissi D, Senhaji N, Aouiss A, Khalki L, Tijani Y, Zaid N, Marhoume FZ, Naya A, Oudghiri M, Kabine M, Zaid Y. IL-1 and CD40/CD40L platelet complex: elements of induction of Crohn's disease and new therapeutic targets. Arch Pharm Res 2021; 44:117-132. [PMID: 33394309 DOI: 10.1007/s12272-020-01296-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 11/21/2020] [Indexed: 12/15/2022]
Abstract
Ulcerative colitis (UC) and Crohn's disease (CD) are chronic and multifactorial diseases that affect the intestinal tract, both characterized by recurrent inflammation of the intestinal mucosa, resulting in abdominal pain, diarrhea, vomiting and, rectal bleeding. Inflammatory bowel diseases (IBD) regroup these two disorders. The exact pathological mechanism of IBD remains ambiguous and poorly known. In genetically predisposed patients, defects in intestinal mucosal barrier are due to an uncontrolled inflammatory response to normal flora. In addition to the genetic predisposition, these defects could be triggered by environmental factors or by a specific lifestyle which is widely accepted as etiological hypothesis. The involvement of the CD40/CD40L platelet complex in the development of IBD has been overwhelmingly demonstrated. CD40L is climacteric in cell signalling in innate and adaptive immunity, the CD40L expression on the platelet cell surface gives them an immunological competence. The IL-1, a major inflammation mediator could be involved in different ways in the development of IBD. Here, we provide a comprehensive review regarding the role of platelet CD40/CD40L in the pathophysiological effect of IL-1 in the development of Crohn's disease (CD). This review could potentially help future approaches aiming to target these two pathways for therapeutic purposes and elucidate the immunological mechanisms driving gut inflammation.
Collapse
Affiliation(s)
- Doha Anka Idrissi
- Department of Biology, Faculty of Sciences, Hassan II University, Casablanca, Morocco
| | - Nezha Senhaji
- Laboratory of Genetic and Molecular Pathology, Faculty of Medicine, Hassan II University, Casablanca, Morocco
| | - Asmae Aouiss
- Department of Biology, Faculty of Sciences, Hassan II University, Casablanca, Morocco
| | - Loubna Khalki
- Faculty of Medicine, Mohammed VI University of Health Sciences (UM6SS), Casablanca, Morocco
| | - Youssef Tijani
- Faculty of Medicine, Mohammed VI University of Health Sciences (UM6SS), Casablanca, Morocco
| | - Nabil Zaid
- Faculty of Sciences, Department of Biology, Mohammed V University, Rabat, Morocco
| | - Fatima Zahra Marhoume
- Faculty of Sciences and Technology, Laboratory of Biochemistry and Neuroscience, Integrative and Computational Neuroscience Team, Hassan First University, Settat, Morocco
| | - Abdallah Naya
- Department of Biology, Faculty of Sciences, Hassan II University, Casablanca, Morocco
| | - Mounia Oudghiri
- Department of Biology, Faculty of Sciences, Hassan II University, Casablanca, Morocco
| | - Mostafa Kabine
- Department of Biology, Faculty of Sciences, Hassan II University, Casablanca, Morocco
| | - Younes Zaid
- Faculty of Sciences, Department of Biology, Mohammed V University, Rabat, Morocco. .,Research Center of Abulcasis, University of Health Sciences, Rabat, Morocco.
| |
Collapse
|
63
|
Campbell RA, Boilard E, Rondina MT. Is there a role for the ACE2 receptor in SARS-CoV-2 interactions with platelets? J Thromb Haemost 2021; 19:46-50. [PMID: 33119197 PMCID: PMC7899240 DOI: 10.1111/jth.15156] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/12/2020] [Accepted: 10/26/2020] [Indexed: 01/11/2023]
Abstract
There is an urgent need to understand the underlying mechanisms contributing to thrombotic and inflammatory complications during COVID-19. Data from independent groups have identified that platelets are hyperreactive during COVID-19. Platelet hyperreactivity is accompanied by changes in platelet gene expression, and enhanced interactions between platelets and leukocytes. In some patients, SARS-CoV-2 mRNA has been detected in platelets. Together, this suggests that SARS-CoV-2 may interact with platelets. However, controversy remains on which receptors mediate SARS-CoV-2 platelet interactions. Most, but not all, transcriptomic and proteomic analyses fail to observe the putative SARS-CoV-2 receptor, angiotensin converting enzyme-2, or the cellular serine protease necessary for viral entry, TMPRSS2, on platelets and megakaryocytes. Interestingly, platelets express other known SARS-CoV-2 receptors, which induce similar patterns of activation to those observed when platelets are incubated with SARS-CoV-2. This article explores these findings and discusses ongoing areas of controversy and uncertainty with regard to SARS-CoV-2 platelet interactions.
Collapse
Affiliation(s)
- Robert A Campbell
- University of Utah Molecular Medicine Program, Salt Lake City, UT, USA
- Departments of Internal Medicine & Pathology, University of Utah, Salt Lake City, UT, USA
| | - Eric Boilard
- Centre de Recherche du Centre Hospitalier,, Universitaire de Québec- Université Laval, Québec, QC, Canada
- Département de Microbiologie-Infectiologie et d'immunologie, Université Laval, Québec, QC, Canada
| | - Matthew T Rondina
- University of Utah Molecular Medicine Program, Salt Lake City, UT, USA
- Departments of Internal Medicine & Pathology, University of Utah, Salt Lake City, UT, USA
- George E. Wahlen VAMC Department of Internal Medicine and GRECC, Salt Lake City, UT, USA
| |
Collapse
|
64
|
The Clot Thickens: Recent Clues on Hematopoietic Stem Cell Contribution to Age-Related Platelet Biology Open New Questions. ADVANCES IN GERIATRIC MEDICINE AND RESEARCH 2021; 3. [PMID: 35037001 PMCID: PMC8759758 DOI: 10.20900/agmr20210019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Platelets provide life-saving functions by halting external and internal bleeding. There is also a dark side to platelet biology, however. Recent reports provide evidence for increased platelet reactivity during aging of mice and humans, making platelets main suspects in the most prevalent aging-related human pathologies, including cardiovascular diseases, stroke, and cancer. What drives this platelet hyperreactivity during aging? Here, we discuss how hematopoietic stem cell differentiation pathways into the platelet lineage offer avenues to understand the fundamental differences between young and old platelets. Recent advances begin to unravel how the cellular and molecular regulation of the parent hematopoietic stem and progenitor cells likely imbue aging characteristics on the resulting Plt progeny. The resulting mechanistic insights into intrinsic platelet reactivity will provide strategies for selectively targeting age-related pathways. This brief viewpoint focuses on current concepts on aging hematopoiesis and the implications for platelet hyperactivity during aging.
Collapse
|
65
|
Danese E, Montagnana M, Gelati M, Lippi G. The Role of Epigenetics in the Regulation of Hemostatic Balance. Semin Thromb Hemost 2020; 47:53-62. [PMID: 33368118 DOI: 10.1055/s-0040-1718400] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Epigenetics, a term conventionally used to explain the intricate interplay between genes and the environment, is now regarded as the fundament of developmental biology. Several lines of evidence garnered over the past decades suggest that epigenetic alterations, mostly encompassing DNA methylation, histone tail modifications, and generation of microRNAs, play an important, though still incompletely explored, role in both primary and secondary hemostasis. Epigenetic variations may interplay with platelet functions and their responsiveness to antiplatelet drugs, and they may also exert a substantial contribution in modulating the production and release into the bloodstream of proteins involved in blood coagulation and fibrinolysis. This emerging evidence may have substantial biological and clinical implications. An enhanced understanding of posttranscriptional mechanisms would help to clarify some remaining enigmatic issues in primary and secondary hemostasis, which cannot be thoughtfully explained by genetics or biochemistry alone. Increased understanding would also pave the way to developing innovative tests for better assessment of individual risk of bleeding or thrombosis. The accurate recognition of key epigenetic mechanisms in hemostasis would then contribute to identify new putative therapeutic targets, and develop innovative agents that could be helpful for preventing or managing a vast array of hemostasis disturbances.
Collapse
Affiliation(s)
- Elisa Danese
- Section of Clinical Biochemistry, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Martina Montagnana
- Section of Clinical Biochemistry, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Matteo Gelati
- Section of Clinical Biochemistry, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Giuseppe Lippi
- Section of Clinical Biochemistry, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| |
Collapse
|
66
|
Kalev-Zylinska ML, Morel-Kopp MC, Ward CM, Hearn JI, Hamilton JR, Bogdanova AY. Ionotropic glutamate receptors in platelets: opposing effects and a unifying hypothesis. Platelets 2020; 32:998-1008. [PMID: 33284715 DOI: 10.1080/09537104.2020.1852542] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Ionotropic glutamate receptors include α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPAR), kainate receptors (KAR), and N-methyl-D-aspartate receptors (NMDAR). All function as cation channels; AMPAR and KAR are more permeable to sodium and NMDAR to calcium ions. Compared to the brain, receptor assemblies in platelets are unusual, suggesting distinctive functionalities.There is convincing evidence that AMPAR and KAR amplify platelet function and thrombus formation in vitro and in vivo. Transgenic mice lacking GluA1 and GluK2 (AMPAR and KAR subunits, respectively) have longer bleeding times and prolonged time to thrombosis in an arterial model. In humans, rs465566 KAR gene polymorphism associates with altered in vitro platelet responses suggesting enhanced aspirin effect. The NMDAR contribution to platelet function is less well defined. NMDA at low concentrations (≤10 μM) inhibits platelet aggregation and high concentrations (≥100 μM) have no effect. However, open NMDAR channel blockers interfere with platelet activation and aggregation induced by other agonists in vitro; anti-GluN1 antibodies interfere with thrombus formation under high shear rates ex vivo; and rats vaccinated with GluN1 develop iron deficiency anemia suggestive of mild chronic bleeding. In this review, we summarize data on glutamate receptors in platelets and propose a unifying model that reconciles some of the opposing effects observed.
Collapse
Affiliation(s)
- Maggie L Kalev-Zylinska
- Blood and Cancer Biology Laboratory, Department of Molecular Medicine & Pathology, University of Auckland, Auckland, New Zealand.,Department of Pathology and Laboratory Medicine, LabPlus Haematology, Auckland City Hospital, Auckland, New Zealand
| | - Marie-Christine Morel-Kopp
- Department of Haematology and Transfusion Medicine, Royal North Shore Hospital, Sydney, Australia.,Northern Blood Research Centre, Kolling Institute, University of Sydney, Sydney, Australia
| | - Christopher M Ward
- Department of Haematology and Transfusion Medicine, Royal North Shore Hospital, Sydney, Australia.,Northern Blood Research Centre, Kolling Institute, University of Sydney, Sydney, Australia
| | - James I Hearn
- Blood and Cancer Biology Laboratory, Department of Molecular Medicine & Pathology, University of Auckland, Auckland, New Zealand
| | - Justin R Hamilton
- Australian Centre for Blood Diseases, Monash University, Melbourne, Australia
| | - Anna Y Bogdanova
- Red Blood Cell Research Group, Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zürich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Zürich, Switzerland
| |
Collapse
|
67
|
Faria AVS, Andrade SS, Peppelenbosch MP, Ferreira-Halder CV, Fuhler GM. Platelets in aging and cancer-"double-edged sword". Cancer Metastasis Rev 2020; 39:1205-1221. [PMID: 32869161 PMCID: PMC7458881 DOI: 10.1007/s10555-020-09926-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 08/12/2020] [Indexed: 02/06/2023]
Abstract
Platelets control hemostasis and play a key role in inflammation and immunity. However, platelet function may change during aging, and a role for these versatile cells in many age-related pathological processes is emerging. In addition to a well-known role in cardiovascular disease, platelet activity is now thought to contribute to cancer cell metastasis and tumor-associated venous thromboembolism (VTE) development. Worldwide, the great majority of all patients with cardiovascular disease and some with cancer receive anti-platelet therapy to reduce the risk of thrombosis. However, not only do thrombotic diseases remain a leading cause of morbidity and mortality, cancer, especially metastasis, is still the second cause of death worldwide. Understanding how platelets change during aging and how they may contribute to aging-related diseases such as cancer may contribute to steps taken along the road towards a "healthy aging" strategy. Here, we review the changes that occur in platelets during aging, and investigate how these versatile blood components contribute to cancer progression.
Collapse
Affiliation(s)
- Alessandra V S Faria
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center Rotterdam, NL-3000 CA, Rotterdam, The Netherlands
- Department of Biochemistry and Tissue Biology, University of Campinas, UNICAMP, Campinas, SP, 13083-862, Brazil
| | | | - Maikel P Peppelenbosch
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center Rotterdam, NL-3000 CA, Rotterdam, The Netherlands
| | - Carmen V Ferreira-Halder
- Department of Biochemistry and Tissue Biology, University of Campinas, UNICAMP, Campinas, SP, 13083-862, Brazil
| | - Gwenny M Fuhler
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center Rotterdam, NL-3000 CA, Rotterdam, The Netherlands.
| |
Collapse
|
68
|
K. Poddar M, Banerjee S. Molecular Aspects of Pathophysiology of Platelet Receptors. Platelets 2020. [DOI: 10.5772/intechopen.92856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Receptor is a dynamic instrumental surface protein that helps to interact with specific molecules to respond accordingly. Platelet is the smallest in size among the blood components, but it plays many pivotal roles to maintain hemostasis involving its surface receptors. It (platelet) has cell adhesion receptors (e.g., integrins and glycoproteins), leucine-rich repeats receptors (e.g., TLRs, glycoprotein complex, and MMPs), selectins (e.g., CLEC, P-selectin, and CD), tetraspanins (e.g., CD and LAMP), transmembrane receptors (e.g., purinergic—P2Y and P2X1), prostaglandin receptors (e.g., TxA2, PGH2, and PGI2), immunoglobulin superfamily receptors (e.g., FcRγ and FcεR), etc. on its surface. The platelet receptors (e.g., glycoproteins, protease-activated receptors, and GPCRs) during platelet activation are over expressed and their granule contents are secreted (including neurotransmitters, cytokines, and chemokines) into circulation, which are found to be correlated with different physiological conditions. Interestingly, platelets promote metastasis through circulation protecting from cytolysis and endogenous immune surveillance involving several platelets receptors. The updated knowledge about different types of platelet receptors in all probable aspects, including their inter- and intra-signaling mechanisms, are discussed with respect to not only its (platelets) receptor type but also under different pathophysiological conditions.
Collapse
|
69
|
Águila S, Cuenca-Zamora E, Martínez C, Teruel-Montoya R. MicroRNAs in Platelets: Should I Stay or Should I Go? Platelets 2020. [DOI: 10.5772/intechopen.93181] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In this chapter, we discuss different topics always using the microRNA as the guiding thread of the review. MicroRNAs, member of small noncoding RNAs family, are an important element involved in gene expression. We cover different issues such as their importance in the differentiation and maturation of megakaryocytes (megakaryopoiesis), as well as the role in platelets formation (thrombopoiesis) focusing on the described relationship between miRNA and critical myeloid lineage transcription factors such as RUNX1, chemokines receptors as CRCX4, or central hormones in platelet homeostasis like TPO, as well as its receptor (MPL) and the TPO signal transduction pathway, that is JAK/STAT. In addition to platelet biogenesis, we review the microRNA participation in platelets physiology and function. This review also introduces the use of miRNAs as biomarkers of platelet function since the detection of pathogenic situations or response to therapy using these noncoding RNAs is getting increasing interest in disease management. Finally, this chapter describes the participation of platelets in cellular interplay, since extracellular vesicles have been demonstrated to have the ability to deliver microRNAs to others cells, modulating their function through intercellular communication, redefining the extracellular vesicles from the so-called “platelet dust” to become mediators of intercellular communication.
Collapse
|
70
|
Untying knots to make more platelets. Blood 2020; 136:1702-1703. [PMID: 33031526 DOI: 10.1182/blood.2020007381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
71
|
Bhatlekar S, Manne BK, Basak I, Edelstein LC, Tugolukova E, Stoller ML, Cody MJ, Morley SC, Nagalla S, Weyrich AS, Rowley JW, O'Connell RM, Rondina MT, Campbell RA, Bray PF. miR-125a-5p regulates megakaryocyte proplatelet formation via the actin-bundling protein L-plastin. Blood 2020; 136:1760-1772. [PMID: 32844999 PMCID: PMC7544541 DOI: 10.1182/blood.2020005230] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 05/24/2020] [Indexed: 12/17/2022] Open
Abstract
There is heritability to interindividual variation in platelet count, and better understanding of the regulating genetic factors may provide insights for thrombopoiesis. MicroRNAs (miRs) regulate gene expression in health and disease, and megakaryocytes (MKs) deficient in miRs have lower platelet counts, but information about the role of miRs in normal human MK and platelet production is limited. Using genome-wide miR profiling, we observed strong correlations among human bone marrow MKs, platelets, and differentiating cord blood-derived MK cultures, and identified MK miR-125a-5p as associated with human platelet number but not leukocyte or hemoglobin levels. Overexpression and knockdown studies showed that miR-125a-5p positively regulated human MK proplatelet (PP) formation in vitro. Inhibition of miR-125a-5p in vivo lowered murine platelet counts. Analyses of MK and platelet transcriptomes identified LCP1 as a miR-125a-5p target. LCP1 encodes the actin-bundling protein, L-plastin, not previously studied in MKs. We show that miR-125a-5p directly targets and reduces expression of MK L-plastin. Overexpression and knockdown studies show that L-plastin promotes MK progenitor migration, but negatively correlates with human platelet count and inhibits MK PP formation (PPF). This work provides the first evidence for the actin-bundling protein, L-plastin, as a regulator of human MK PPF via inhibition of the late-stage MK invagination system, podosome and PPF, and PP branching. We also provide resources of primary and differentiating MK transcriptomes and miRs associated with platelet counts. miR-125a-5p and L-plastin may be relevant targets for increasing in vitro platelet manufacturing and for managing quantitative platelet disorders.
Collapse
Affiliation(s)
- Seema Bhatlekar
- Program in Molecular Medicine, University of Utah, Salt Lake City, UT
| | - Bhanu K Manne
- Program in Molecular Medicine, University of Utah, Salt Lake City, UT
| | - Indranil Basak
- Program in Molecular Medicine, University of Utah, Salt Lake City, UT
| | - Leonard C Edelstein
- Cardeza Foundation for Hematologic Research, Thomas Jefferson University, Philadelphia, PA
| | - Emilia Tugolukova
- Program in Molecular Medicine, University of Utah, Salt Lake City, UT
| | | | - Mark J Cody
- Program in Molecular Medicine, University of Utah, Salt Lake City, UT
| | - Sharon C Morley
- Division of Infectious Diseases, Department of Pediatrics and
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Srikanth Nagalla
- Division of Hematology and Oncology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX
| | - Andrew S Weyrich
- Program in Molecular Medicine, University of Utah, Salt Lake City, UT
- Division of Pulmonary, Department of Internal Medicine
| | - Jesse W Rowley
- Program in Molecular Medicine, University of Utah, Salt Lake City, UT
- Division of Pulmonary, Department of Internal Medicine
| | - Ryan M O'Connell
- Division of Microbiology and Immunology, Department of Pathology, and
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Matthew T Rondina
- Program in Molecular Medicine, University of Utah, Salt Lake City, UT
- Geriatric Research, Education and Clinical Center, George E. Wahlen VAMC GRECC, Salt Lake City, UT; and
- Division of General Internal Medicine and
| | - Robert A Campbell
- Program in Molecular Medicine, University of Utah, Salt Lake City, UT
- Division of General Internal Medicine and
| | - Paul F Bray
- Program in Molecular Medicine, University of Utah, Salt Lake City, UT
- Division of Hematology and Hematologic Malignancies, Department of Internal Medicine, University of Utah, Salt Lake City, UT
| |
Collapse
|
72
|
Won E, Morodomi Y, Kanaji S, Shapiro R, Vo M, Orje JN, Thornburg CD, Yang X, Ruggeri ZM, Schimmel P, Kanaji T. Extracellular tyrosyl-tRNA synthetase cleaved by plasma proteinases and stored in platelet α-granules: Potential role in monocyte activation. Res Pract Thromb Haemost 2020; 4:1167-1177. [PMID: 33134783 PMCID: PMC7590329 DOI: 10.1002/rth2.12429] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 08/18/2020] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Tyrosyl-tRNA synthetase (YRS) belongs to the family of enzymes that catalyzes the tRNA aminoacylation reaction for protein synthesis, and it has been recently shown to exert noncanonical functions. Although database results indicate extremely low levels of YRS mRNA in platelets, YRS protein is abundantly present. The source of YRS in platelets, as well as the physiological role of platelet-stored YRS, remains largely unknown. OBJECTIVES To clarify how YRS accumulates in platelets and determine the potential role of platelet-stored YRS. METHODS Recombinant YRS proteins with epitope tags were prepared and tested in vitro for proteolytic cleavage in human plasma. Fluorescent-labeled YRS was examined for uptake by platelets, as demonstrated by western blotting and confocal microscopy analysis. Using RAW-Dual reporter cells, Toll-like receptor and type I interferon activation pathways were analyzed after treatment with YRS. RESULTS Full-length YRS was cleaved by both elastase and matrix metalloproteinases in the plasma. The cleaved, N-terminal YRS fragment corresponds to the endogenous YRS detected in platelet lysate by western blotting. Both full-length and cleaved forms of YRS were taken up by platelets in vitro and stored in the α-granules. The N-terminal YRS fragment generated by proteolytic cleavage had monocyte activation comparable to that of the constitutive-active mutant YRS (YRSY341A) previously reported. CONCLUSION Platelets take up both full-length YRS and the active form of cleaved YRS fragment from the plasma. The cleaved, N-terminal YRS fragment stored in α-granules may have potential to activate monocytes.
Collapse
Affiliation(s)
- Eric Won
- Department of Molecular MedicineMERU‐Roon Research Center on Vascular BiologyThe Scripps Research InstituteLa JollaCaliforniaUSA
- Division of Hematology/OncologyDepartment of PediatricsUC San Diego School of MedicineLa JollaCaliforniaUSA
- Hemophilia and Thrombosis Treatment CenterRady Children's HospitalSan DiegoCaliforniaUSA
| | - Yosuke Morodomi
- Department of Molecular MedicineMERU‐Roon Research Center on Vascular BiologyThe Scripps Research InstituteLa JollaCaliforniaUSA
| | - Sachiko Kanaji
- Department of Molecular MedicineMERU‐Roon Research Center on Vascular BiologyThe Scripps Research InstituteLa JollaCaliforniaUSA
- Department of Molecular MedicineThe Scripps Laboratories for tRNA Synthetase ResearchThe Scripps Research InstituteLa JollaCaliforniaUSA
| | - Ryan Shapiro
- Department of Molecular MedicineThe Scripps Laboratories for tRNA Synthetase ResearchThe Scripps Research InstituteLa JollaCaliforniaUSA
| | - My‐Nuong Vo
- Department of Molecular MedicineThe Scripps Laboratories for tRNA Synthetase ResearchThe Scripps Research InstituteLa JollaCaliforniaUSA
| | - Jennifer N. Orje
- Department of Molecular MedicineMERU‐Roon Research Center on Vascular BiologyThe Scripps Research InstituteLa JollaCaliforniaUSA
| | - Courtney D. Thornburg
- Division of Hematology/OncologyDepartment of PediatricsUC San Diego School of MedicineLa JollaCaliforniaUSA
- Hemophilia and Thrombosis Treatment CenterRady Children's HospitalSan DiegoCaliforniaUSA
| | - Xiang‐Lei Yang
- Department of Molecular MedicineThe Scripps Laboratories for tRNA Synthetase ResearchThe Scripps Research InstituteLa JollaCaliforniaUSA
| | - Zaverio M. Ruggeri
- Department of Molecular MedicineMERU‐Roon Research Center on Vascular BiologyThe Scripps Research InstituteLa JollaCaliforniaUSA
| | - Paul Schimmel
- Department of Molecular MedicineThe Scripps Laboratories for tRNA Synthetase ResearchThe Scripps Research InstituteLa JollaCaliforniaUSA
- Department of Molecular MedicineThe Scripps Research InstituteJupiterFloridaUSA
| | - Taisuke Kanaji
- Department of Molecular MedicineMERU‐Roon Research Center on Vascular BiologyThe Scripps Research InstituteLa JollaCaliforniaUSA
| |
Collapse
|
73
|
Fazmin IT, Achercouk Z, Edling CE, Said A, Jeevaratnam K. Circulating microRNA as a Biomarker for Coronary Artery Disease. Biomolecules 2020; 10:E1354. [PMID: 32977454 PMCID: PMC7598281 DOI: 10.3390/biom10101354] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/15/2020] [Accepted: 09/19/2020] [Indexed: 12/14/2022] Open
Abstract
Coronary artery disease (CAD) is the leading cause of sudden cardiac death in adults, and new methods of predicting disease and risk-stratifying patients will help guide intervention in order to reduce this burden. Current CAD detection involves multiple modalities, but the consideration of other biomarkers will help improve reliability. The aim of this narrative review is to help researchers and clinicians appreciate the growing relevance of miRNA in CAD and its potential as a biomarker, and also to suggest useful miRNA that may be targets for future study. We sourced information from several databases, namely PubMed, Scopus, and Google Scholar, when collating evidentiary information. MicroRNAs (miRNA) are short, noncoding RNAs that are relevant in cardiovascular physiology and pathophysiology, playing roles in cardiac hypertrophy, maintenance of vascular tone, and responses to vascular injury. CAD is associated with changes in miRNA expression profiles, and so are its risk factors, such as abnormal lipid metabolism and inflammation. Thus, they may potentially be biomarkers of CAD. Nevertheless, there are limitations in using miRNA. These include cost and the presence of several confounding factors that may affect miRNA profiles. Furthermore, there is difficulty in the normalisation of miRNA values between published studies, due to pre-analytical variations in samples.
Collapse
Affiliation(s)
- Ibrahim T. Fazmin
- Faculty of Health and Medical Science, University of Surrey, Guildford GU2 7AL, UK; (I.T.F.); (Z.A.); (C.E.E.)
- School of Clinical Medicine, University of Cambridge, Cambridge CB2 1TN, UK
| | - Zakaria Achercouk
- Faculty of Health and Medical Science, University of Surrey, Guildford GU2 7AL, UK; (I.T.F.); (Z.A.); (C.E.E.)
| | - Charlotte E. Edling
- Faculty of Health and Medical Science, University of Surrey, Guildford GU2 7AL, UK; (I.T.F.); (Z.A.); (C.E.E.)
| | - Asri Said
- School of Medicine, University Malaysia Sarawak, Kota Samarahan 94300, Sarawak, Malaysia;
| | - Kamalan Jeevaratnam
- Faculty of Health and Medical Science, University of Surrey, Guildford GU2 7AL, UK; (I.T.F.); (Z.A.); (C.E.E.)
| |
Collapse
|
74
|
Esparza O, Higa K, Davizon-Castillo P. Molecular and functional characteristics of megakaryocytes and platelets in aging. Curr Opin Hematol 2020; 27:302-310. [PMID: 32740036 PMCID: PMC11776438 DOI: 10.1097/moh.0000000000000601] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Advances in medical care and preventive measures have contributed to increasing life expectancy. Therefore, it is critical to expand our understanding of the physiological and pathophysiological adaptations of the hematological system in aging. We highlight and review the findings from recent investigations aimed at understanding the effects of aging on megakaryocytes and platelets. RECENT FINDINGS Biochemical and transcriptomic studies of megakaryocytes and platelets from older humans and mice have advanced our understanding of the molecular and functional characteristics of megakaryocytes and platelets during aging. These studies have led to the identification of metabolic and inflammatory pathways associated with the generation of hyperreactive platelets that may significantly contribute to the high incidence of thrombosis in aging. SUMMARY By increasing our research efforts to understand and identify the characteristics of megakaryocytes and platelets in aging, we will increase our potential to develop novel therapies aimed at decreasing the incidence of aging-associated thrombosis. These efforts will also serve as a foundation to better understand the role of megakaryocytes and platelets in other age-related hematological conditions with high thrombotic risk such as clonal hematopoiesis of indeterminate potential and myeloproliferative neoplasms.
Collapse
Affiliation(s)
- Orlando Esparza
- Department of Pediatric Hematology, Oncology, and Bone Marrow Transplant, University of Colorado, Aurora, Colorado, USA
| | - Kelly Higa
- Medical Scientist Training Program, University of Colorado, Aurora, Colorado, USA
| | - Pavel Davizon-Castillo
- Department of Pediatric Hematology, Oncology, and Bone Marrow Transplant, University of Colorado, Aurora, Colorado, USA
- Hemophilia and Thrombosis Center, School of Medicine, University of Colorado, Aurora, Colorado, USA
| |
Collapse
|
75
|
Patel P, Naik UP. Platelet MAPKs-a 20+ year history: What do we really know? J Thromb Haemost 2020; 18:2087-2102. [PMID: 32574399 DOI: 10.1111/jth.14967] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/09/2020] [Accepted: 06/10/2020] [Indexed: 01/01/2023]
Abstract
The existence of mitogen activated protein kinases (MAPKs) in platelets has been known for more than 20 years. Since that time hundreds of reports have been published describing the conditions that cause MAPK activation in platelets and their role in regulating diverse platelet functions from the molecular to physiological level. However, this cacophony of reports, with inconsistent and sometimes contradictory findings, has muddied the waters leading to great confusion. Since the last review of platelet MAPKs was published more than a decade ago, there have been more than 50 reports, including the description of novel knockout mouse models, that have furthered our knowledge. Therefore, we undertook an extensive literature review to delineate what is known about platelet MAPKs. We specifically discuss what is currently known about how MAPKs are activated and what signaling cascades they regulate in platelets incorporating recent findings from knockout mouse models. In addition, we will discuss the role each MAPK plays in regulating distinct platelet functions. In doing so, we hope to clarify the role for MAPKs and identify knowledge gaps in this field that await future researchers. In addition, we discuss the limitations of current studies with a particular focus on the off-target effects of commonly used MAPK inhibitors. We conclude with a look at the clinical utility of MAPK inhibitors as potential antithrombotic therapies with an analysis of current clinical trial data.
Collapse
Affiliation(s)
- Pravin Patel
- Department of Medicine, Cardeza Center for Hemostasis, Thrombosis, and Vascular Biology, Cardeza Foundation for Hematologic Research, Thomas Jefferson University, Philadelphia, PA, USA
| | - Ulhas P Naik
- Department of Medicine, Cardeza Center for Hemostasis, Thrombosis, and Vascular Biology, Cardeza Foundation for Hematologic Research, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
76
|
Lazar S, Wurtzel JGT, Chen X, Ma P, Goldfinger LE. High-efficiency unassisted transfection of platelets with naked double-stranded miRNAs modulates signal-activated translation and platelet function. Platelets 2020; 32:794-806. [PMID: 32838617 DOI: 10.1080/09537104.2020.1809642] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
We sought novel approaches to improve transfection efficiencies of microRNAs (miRNAs) in platelets, and to apply these approaches to investigate the roles of miRNAs in regulating signal-activated protein translation and functional effects. We found that ex vivo human platelets support gymnosis---internalization of ectopic miRNAs following co-incubation in the absence of conventional transfection reagents or schemes---and subsequently incorporate transfected miRNA into ARGONAUTE2 (AGO2)-based RNA-induced silencing complexes (RISC). Thrombin/fibrinogen stimulation activated translation of miR-223-3p target SEPTIN2, which was suppressed by miR-223-3p transfection in an AGO2/RISC-dependent manner. Thrombin/fibrinogen-induced exosome and microvesicle generation was inhibited by miR-223-3p transfection, and this effect was reversed with a RISC inhibitor. Platelet gymnosis of naked miRNAs appeared to be mediated in part by endocytic pathways including clathrin-dependent and fluid-phase endocytosis and caveolae. These results demonstrate the ability of ex vivo platelets to internalize ectopic miRNAs by unassisted transfection, and utilize them to modulate signal-activated translation and platelet function. Our results identify new roles for miR-223-3p in extracellular vesicle generation in stimulated platelets. High-efficiency gymnotic transfection of miRNAs in ex vivo platelets may be a broadly useful tool for exploring molecular genetic regulation of platelet function.
Collapse
Affiliation(s)
- Sophia Lazar
- Cardeza Foundation for Hematologic Research, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Jeremy G T Wurtzel
- Cardeza Foundation for Hematologic Research, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Xi Chen
- Cardeza Foundation for Hematologic Research, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Peisong Ma
- Cardeza Foundation for Hematologic Research, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Lawrence E Goldfinger
- Cardeza Foundation for Hematologic Research, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
77
|
Rodriguez BA, Bhan A, Beswick A, Elwood PC, Niiranen TJ, Salomaa V, Trégouët DA, Morange PE, Civelek M, Ben-Shlomo Y, Schlaeger T, Chen MH, Johnson AD, Johnson AD. A Platelet Function Modulator of Thrombin Activation Is Causally Linked to Cardiovascular Disease and Affects PAR4 Receptor Signaling. Am J Hum Genet 2020; 107:211-221. [PMID: 32649856 DOI: 10.1016/j.ajhg.2020.06.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 06/03/2020] [Indexed: 12/17/2022] Open
Abstract
Dual antiplatelet therapy reduces ischemic events in cardiovascular disease, but it increases bleeding risk. Thrombin receptors PAR1 and PAR4 are drug targets, but the role of thrombin in platelet aggregation remains largely unexplored in large populations. We performed a genome-wide association study (GWAS) of platelet aggregation in response to full-length thrombin, followed by clinical association analyses, Mendelian randomization, and functional characterization including iPSC-derived megakaryocyte and platelet experiments. We identified a single sentinel variant in the GRK5 locus (rs10886430-G, p = 3.0 × 10-42) associated with increased thrombin-induced platelet aggregation (β = 0.70, SE = 0.05). We show that disruption of platelet GRK5 expression by rs10886430-G is associated with enhanced platelet reactivity. The proposed mechanism of a GATA1-driven megakaryocyte enhancer is confirmed in allele-specific experiments. Utilizing further data, we demonstrate that the allelic effect is highly platelet- and thrombin-specific and not likely due to effects on thrombin levels. The variant is associated with increased risk of cardiovascular disease outcomes in UK BioBank, most strongly with pulmonary embolism. The variant associates with increased risk of stroke in the MEGASTROKE, UK BioBank, and FinnGen studies. Mendelian randomization analyses in independent samples support a causal role for rs10886430-G in increasing risk for stroke, pulmonary embolism, and venous thromboembolism through its effect on thrombin-induced platelet reactivity. We demonstrate that G protein-coupled receptor kinase 5 (GRK5) promotes platelet activation specifically via PAR4 receptor signaling. GRK5 inhibitors in development for the treatment of heart failure and cancer could have platelet off-target deleterious effects. Common variants in GRK5 may modify clinical outcomes with PAR4 inhibitors, and upregulation of GRK5 activity or signaling in platelets may have therapeutic benefits.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Andrew D Johnson
- National Heart, Lung, and Blood Institute, Division of Intramural Research, Population Sciences Branch, The Framingham Heart Study, Framingham, MA 01702, USA.
| |
Collapse
|
78
|
Chicharro P, Rodríguez-Jiménez P, Llamas-Velasco M, Montes N, Sanz-García A, Cibrian D, Vara A, Gómez MJ, Jiménez-Fernández M, Martínez-Fleta P, Sánchez-García I, Lozano-Prieto M, Triviño JC, Miñambres R, Sánchez-Madrid F, de la Fuente H, Dauden E. Expression of miR-135b in Psoriatic Skin and Its Association with Disease Improvement. Cells 2020; 9:cells9071603. [PMID: 32630692 PMCID: PMC7408353 DOI: 10.3390/cells9071603] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 06/29/2020] [Accepted: 06/30/2020] [Indexed: 12/17/2022] Open
Abstract
miRNAs have been associated with psoriasis since just over a decade. However, we are far from a complete understanding of their role during the development of this disease. Our objective was to characterize the cutaneous expression of miRNAs not previously described in psoriasis, the changes induced following the treatment with biologicals and their association with disease improvement. Next generation sequencing was performed from five skin samples from psoriasis patients (lesional and non-lesional skin) and five controls, and from this cohort, 12 microRNAs were selected to be analyzed in skin samples from 44 patients with plaque psoriasis. In 15 patients, an additional sample was obtained after three months of biological treatment. MiR-9-5p, miR-133a-3p and miR-375 were downregulated in the lesional skin of psoriasis patients. After treatment, expression of miR-133a-3p, miR-375, miR-378a and miR-135b in residual lesions returned towards the levels observed in non-lesional skin. The decrease in miR-135b levels after treatment with biologics was associated with both the improvement of patients evaluated through Psoriasis Area and Severity Index score and the decrease in local inflammatory response. Moreover, basal expression of miR-135b along with age was associated with the improvement of psoriasis, suggesting its possible usefulness as a prognostic biomarker.
Collapse
Affiliation(s)
- Pablo Chicharro
- Dermatology Department, Instituto de Investigación Sanitaria Hospital Universitario de la Princesa (IISP), 28006 Madrid, Spain; (P.C.); (P.R.-J.); (M.L.-V.); (E.D.)
| | - Pedro Rodríguez-Jiménez
- Dermatology Department, Instituto de Investigación Sanitaria Hospital Universitario de la Princesa (IISP), 28006 Madrid, Spain; (P.C.); (P.R.-J.); (M.L.-V.); (E.D.)
| | - Mar Llamas-Velasco
- Dermatology Department, Instituto de Investigación Sanitaria Hospital Universitario de la Princesa (IISP), 28006 Madrid, Spain; (P.C.); (P.R.-J.); (M.L.-V.); (E.D.)
| | - Nuria Montes
- Rheumatology Department, Instituto de Investigación Sanitaria Hospital Universitario de la Princesa (IISP), 28006 Madrid, Spain;
- Fisiología Vegetal, Departamento Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, 28003 Madrid, Spain
| | - Ancor Sanz-García
- Data Analysis Unit, Instituto de Investigación Sanitaria Hospital Universitario de la Princesa (IISP), 28006 Madrid, Spain;
| | - Danay Cibrian
- Immunology Department, Instituto de Investigación Sanitaria Hospital Universitario de la Princesa (IISP), 28006 Madrid, Spain; (D.C.); (A.V.); (M.J.-F.); (P.M.-F.); (I.S.-G.); (M.L.-P.); (F.S.-M.)
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, 28009 Madrid, Spain
| | - Alicia Vara
- Immunology Department, Instituto de Investigación Sanitaria Hospital Universitario de la Princesa (IISP), 28006 Madrid, Spain; (D.C.); (A.V.); (M.J.-F.); (P.M.-F.); (I.S.-G.); (M.L.-P.); (F.S.-M.)
| | - Manuel J Gómez
- Bioinformatics Unit, Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain;
| | - María Jiménez-Fernández
- Immunology Department, Instituto de Investigación Sanitaria Hospital Universitario de la Princesa (IISP), 28006 Madrid, Spain; (D.C.); (A.V.); (M.J.-F.); (P.M.-F.); (I.S.-G.); (M.L.-P.); (F.S.-M.)
| | - Pedro Martínez-Fleta
- Immunology Department, Instituto de Investigación Sanitaria Hospital Universitario de la Princesa (IISP), 28006 Madrid, Spain; (D.C.); (A.V.); (M.J.-F.); (P.M.-F.); (I.S.-G.); (M.L.-P.); (F.S.-M.)
| | - Inés Sánchez-García
- Immunology Department, Instituto de Investigación Sanitaria Hospital Universitario de la Princesa (IISP), 28006 Madrid, Spain; (D.C.); (A.V.); (M.J.-F.); (P.M.-F.); (I.S.-G.); (M.L.-P.); (F.S.-M.)
| | - Marta Lozano-Prieto
- Immunology Department, Instituto de Investigación Sanitaria Hospital Universitario de la Princesa (IISP), 28006 Madrid, Spain; (D.C.); (A.V.); (M.J.-F.); (P.M.-F.); (I.S.-G.); (M.L.-P.); (F.S.-M.)
| | - Juan C Triviño
- Sistemas Genómicos, 46980 Valencia, Spain; (J.C.T.); (R.M.)
| | | | - Francisco Sánchez-Madrid
- Immunology Department, Instituto de Investigación Sanitaria Hospital Universitario de la Princesa (IISP), 28006 Madrid, Spain; (D.C.); (A.V.); (M.J.-F.); (P.M.-F.); (I.S.-G.); (M.L.-P.); (F.S.-M.)
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, 28009 Madrid, Spain
| | - Hortensia de la Fuente
- Immunology Department, Instituto de Investigación Sanitaria Hospital Universitario de la Princesa (IISP), 28006 Madrid, Spain; (D.C.); (A.V.); (M.J.-F.); (P.M.-F.); (I.S.-G.); (M.L.-P.); (F.S.-M.)
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, 28009 Madrid, Spain
- Correspondence:
| | - Esteban Dauden
- Dermatology Department, Instituto de Investigación Sanitaria Hospital Universitario de la Princesa (IISP), 28006 Madrid, Spain; (P.C.); (P.R.-J.); (M.L.-V.); (E.D.)
| |
Collapse
|
79
|
Ghazi AR, Kong X, Chen ES, Edelstein LC, Shaw CA. Bayesian modelling of high-throughput sequencing assays with malacoda. PLoS Comput Biol 2020; 16:e1007504. [PMID: 32692749 PMCID: PMC7394446 DOI: 10.1371/journal.pcbi.1007504] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 07/31/2020] [Accepted: 06/09/2020] [Indexed: 12/13/2022] Open
Abstract
NGS studies have uncovered an ever-growing catalog of human variation while leaving an enormous gap between observed variation and experimental characterization of variant function. High-throughput screens powered by NGS have greatly increased the rate of variant functionalization, but the development of comprehensive statistical methods to analyze screen data has lagged. In the massively parallel reporter assay (MPRA), short barcodes are counted by sequencing DNA libraries transfected into cells and the cell's output RNA in order to simultaneously measure the shifts in transcription induced by thousands of genetic variants. These counts present many statistical challenges, including overdispersion, depth dependence, and uncertain DNA concentrations. So far, the statistical methods used have been rudimentary, employing transformations on count level data and disregarding experimental and technical structure while failing to quantify uncertainty in the statistical model. We have developed an extensive framework for the analysis of NGS functionalization screens available as an R package called malacoda (available from github.com/andrewGhazi/malacoda). Our software implements a probabilistic, fully Bayesian model of screen data. The model uses the negative binomial distribution with gamma priors to model sequencing counts while accounting for effects from input library preparation and sequencing depth. The method leverages the high-throughput nature of the assay to estimate the priors empirically. External annotations such as ENCODE data or DeepSea predictions can also be incorporated to obtain more informative priors-a transformative capability for data integration. The package also includes quality control and utility functions, including automated barcode counting and visualization methods. To validate our method, we analyzed several datasets using malacoda and alternative MPRA analysis methods. These data include experiments from the literature, simulated assays, and primary MPRA data. We also used luciferase assays to experimentally validate several hits from our primary data, as well as variants for which the various methods disagree and variants detectable only with the aid of external annotations.
Collapse
Affiliation(s)
- Andrew R. Ghazi
- Quantitative and Computational Biosciences, Baylor College of Medicine, Houston, Texas, United States of America
| | - Xianguo Kong
- Cardeza Foundation for Hematologic Research, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Ed S. Chen
- Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Leonard C. Edelstein
- Cardeza Foundation for Hematologic Research, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Chad A. Shaw
- Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| |
Collapse
|
80
|
Whole blood transcriptome profile at hospital admission discriminates between patients with ST-segment elevation and non-ST-segment elevation acute myocardial infarction. Sci Rep 2020; 10:8731. [PMID: 32457432 PMCID: PMC7250845 DOI: 10.1038/s41598-020-65527-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 05/06/2020] [Indexed: 12/20/2022] Open
Abstract
Whether ST-segment (STEMI) and non-ST-segment elevation myocardial infarction (NSTEMI) should be regarded as distinct pathophysiological entities is a matter of debate. We tested the hypothesis that peripheral blood gene-expression profiles at presentation distinguish STEMI from NSTEMI. We performed a case-control study collecting whole-blood from 60 STEMI and 58 NSTEMI (defined according to the third universal definition of MI) consecutive patients on hospital admission. We used RNA-sequencing for the discovery phase, comparing 15 STEMI vs. 15 NSTEMI patients, matched for age, sex, and cardiovascular risk factors, and quantitative PCR in the remaining unmatched patients for validating top-significant genes. Gene-level differential expression analysis identified significant differences in the expression of 323 genes: 153 genes withstood correction for admission cardiac troponin I (cTnI), differentiating the two conditions independently of myocardial necrosis extent. Functional annotation analysis uncovered divergent modulation in leukocyte and platelet activation, cell migration, and mitochondrial respiratory processes. Linear regression analysis revealed gene expression patterns on admission predicting infarct size, as indexed by cTnI peak (R2 = 0.58–0.75). Our results unveil distinctive pathological traits for these two MI subtypes and provide insights into the early assessment of injury extent. This could translate into RNA-based disease-specific biomarkers for precision diagnosis and risk stratification.
Collapse
|
81
|
Kenny M, Schoen I. A handshake between platelets and neutrophils might fuel deep vein thrombosis. Platelets 2020; 31:624-626. [DOI: 10.1080/09537104.2020.1769053] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Martin Kenny
- School of Pharmacy and Biomolecular Sciences, Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Ingmar Schoen
- School of Pharmacy and Biomolecular Sciences, Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
| |
Collapse
|
82
|
microRNAs as promising biomarkers of platelet activity in antiplatelet therapy monitoring. Int J Mol Sci 2020; 21:ijms21103477. [PMID: 32423125 PMCID: PMC7278969 DOI: 10.3390/ijms21103477] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/08/2020] [Accepted: 05/11/2020] [Indexed: 12/13/2022] Open
Abstract
Given the high morbidity and mortality of cardiovascular diseases (CVDs), novel biomarkers for platelet reactivity are urgently needed. Ischemic events in CVDs are causally linked to platelets, small anucleate cells important for hemostasis. The major side-effect of antiplatelet therapy are life-threatening bleeding events. Current platelet function tests are not sufficient in guiding treatment decisions. Platelets host a broad spectrum of microRNAs (miRNAs) and are a major source of cell-free miRNAs in the blood stream. Platelet-related miRNAs have been suggested as biomarkers of platelet activation and assessment of antiplatelet therapy responsiveness. Platelets release miRNAs upon activation, possibly leading to alterations of plasma miRNA levels in conjunction with CVD or inadequate platelet inhibition. Unlike current platelet function tests, which measure platelet activation ex vivo, signatures of platelet-related miRNAs potentially enable the assessment of in vivo platelet reactivity. Evidence suggests that some miRNAs are responsive to platelet inhibition, making them promising biomarker candidates. In this review, we explain the secretion of miRNAs upon platelet activation and discuss the potential use of platelet-related miRNAs as biomarkers for CVD and antiplatelet therapy monitoring, but also highlight remaining gaps in our knowledge and uncertainties regarding clinical utility. We also elaborate on technical issues and limitations concerning plasma miRNA quantification.
Collapse
|
83
|
Davizon-Castillo P, Rowley JW, Rondina MT. Megakaryocyte and Platelet Transcriptomics for Discoveries in Human Health and Disease. Arterioscler Thromb Vasc Biol 2020; 40:1432-1440. [PMID: 32295424 PMCID: PMC7253186 DOI: 10.1161/atvbaha.119.313280] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Anucleate platelets, long viewed as merely cell fragments with a limited repertoire of rapid-acting hemostatic functions, are now recognized to have a complex and dynamic transcriptome mirroring that of many nucleated cells. The field of megakaryocyte and platelet transcriptomics has been rapidly growing, particularly with the advent of newer technologies such as next-generation RNA-sequencing. Studies interrogating the megakaryocyte and platelet transcriptome have led to a number of key insights into human health and disease. In this brief focused review, we will discuss some of the recent discoveries made through transcriptome analysis of megakaryocytes and platelets. We will also highlight the utility of integrating ribosome footprint analysis to augment discoveries. Both bulk and single-cell sequencing approaches will be reviewed, along with comparative studies between human and murine platelets under basal healthy settings and during acute systemic inflammatory diseases.
Collapse
Affiliation(s)
- Pavel Davizon-Castillo
- From the Section of Pediatric Hematology, Oncology, and Bone Marrow Transplant, University of Colorado, Aurora (P.D.-C)
| | - Jesse W Rowley
- University of Utah Molecular Medicine Program, University of Utah, Salt Lake City (J.W.R., M.T.R.).,Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City (J.W.R., M.T.R.)
| | - Matthew T Rondina
- From the Section of Pediatric Hematology, Oncology, and Bone Marrow Transplant, University of Colorado, Aurora (P.D.-C).,University of Utah Molecular Medicine Program, University of Utah, Salt Lake City (J.W.R., M.T.R.).,Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City (J.W.R., M.T.R.).,Department of Pathology, University of Utah, Salt Lake City (M.T.R.).,George E. Wahlen VAMC, Salt Lake City, UT (M.T.R.)
| |
Collapse
|
84
|
Wang J, Yao Y, Zhang J, Tang X, Meng X, Wang M, Song L, Yuan J. Platelet microRNA-15b protects against high platelet reactivity in patients undergoing percutaneous coronary intervention through Bcl-2-mediated platelet apoptosis. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:364. [PMID: 32355808 PMCID: PMC7186638 DOI: 10.21037/atm.2020.02.88] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Background High platelet reactivity (HPR) and low platelet reactivity (LPR) are associated with an increased risk of ischemic/bleeding events in patients undergoing percutaneous coronary intervention (PCI). The role platelet miRNAs carry out in platelet reactivity regulation is largely unknown. Methods In this study, we profiled the expression pattern of platelet miRNA in patients undergoing PCI with HPR (n=4) and LPR (n=4) by miRNA microarray screening. The candidate miRNAs were further validated in a larger sample of 17 LPR and 22 HPR patients by quantitative reverse-transcription polymerase chain reaction (RT-qPCR), and miR-15b was found differentially expressed. MiR-15b mimic and inhibitor were transfected into MEG-01 cells, then Bcl-2 protein expression and cell apoptosis were assessed. The relationship between platelet reactivity and platelet apoptosis was further evaluated. ABT-737, a Bcl-2 inhibitor was used to induce platelet apoptosis in PCI patients in vitro, and the influence of enhanced platelet apoptosis on platelet reactivity was explored. Results Two miRNAs were found to be differentially expressed in patients with LPR and HPR using microarray system. Furthermore, the expression of miR-15b, a miRNA known to induce cell apoptosis via targeting of Bcl-2, was confirmed by RT-qPCR (P=0.020) to be 1.4× higher in the platelets of LPR patients than in those of HPR patients. Overexpression of miR-15b was demonstrated to suppress Bcl-2 protein expression and enhance cell apoptosis in a megakaryocyte cell line (MEG-01). The platelets of LPR patients expressed lower levels of Bcl-2 protein than those of HPR patients, and an inverse relationship between platelet reactivity and platelet apoptosis was observed among 44 patients who underwent PCI. Inducing platelet apoptosis in PCI patients in vitro, we observed that their platelet reactivity was decreased in a dose-dependent manner. Conclusions Through the promotion of platelet apoptosis, platelet miR-15b negatively regulates platelet reactivity in patients undergoing PCI. Platelet apoptosis may represent a novel antiplatelet target for overcoming HPR in PCI treatment.
Collapse
Affiliation(s)
- Jinghan Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Yi Yao
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Jiahui Zhang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Xiaofang Tang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Xianmin Meng
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Miao Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Lei Song
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Jinqing Yuan
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| |
Collapse
|
85
|
Rondina MT, Voora D, Simon LM, Schwertz H, Harper JF, Lee O, Bhatlekar SC, Li Q, Eustes AS, Montenont E, Campbell RA, Tolley ND, Kosaka Y, Weyrich AS, Bray PF, Rowley JW. Longitudinal RNA-Seq Analysis of the Repeatability of Gene Expression and Splicing in Human Platelets Identifies a Platelet SELP Splice QTL. Circ Res 2020; 126:501-516. [PMID: 31852401 PMCID: PMC7323475 DOI: 10.1161/circresaha.119.315215] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 12/18/2019] [Indexed: 12/16/2022]
Abstract
RATIONALE Longitudinal studies are required to distinguish within versus between-individual variation and repeatability of gene expression. They are uniquely positioned to decipher genetic signal from environmental noise, with potential application to gene variant and expression studies. However, longitudinal analyses of gene expression in healthy individuals-especially with regards to alternative splicing-are lacking for most primary cell types, including platelets. OBJECTIVE To assess repeatability of gene expression and splicing in platelets and use repeatability to identify novel platelet expression quantitative trait loci (QTLs) and splice QTLs. METHODS AND RESULTS We sequenced the transcriptome of platelets isolated repeatedly up to 4 years from healthy individuals. We examined within and between individual variation and repeatability of platelet RNA expression and exon skipping, a readily measured alternative splicing event. We find that platelet gene expression is generally stable between and within-individuals over time-with the exception of a subset of genes enriched for the inflammation gene ontology. We show an enrichment among repeatable genes for associations with heritable traits, including known and novel platelet expression QTLs. Several exon skipping events were also highly repeatable, suggesting heritable patterns of splicing in platelets. One of the most repeatable was exon 14 skipping of SELP. Accordingly, we identify rs6128 as a platelet splice QTL and define an rs6128-dependent association between SELP exon 14 skipping and race. In vitro experiments demonstrate that this single nucleotide variant directly affects exon 14 skipping and changes the ratio of transmembrane versus soluble P-selectin protein production. CONCLUSIONS We conclude that the platelet transcriptome is generally stable over 4 years. We demonstrate the use of repeatability of gene expression and splicing to identify novel platelet expression QTLs and splice QTLs. rs6128 is a platelet splice QTL that alters SELP exon 14 skipping and soluble versus transmembrane P-selectin protein production.
Collapse
Affiliation(s)
- Matthew T. Rondina
- Molecular Medicine Program
- Department of Internal Medicine
- George E. Wahlen VAMC Geriatric Research and Education Clinical Center, The University of Utah
| | - Deepak Voora
- Duke Center for Applied Genomics & Precision Medicine, Durham, NC
| | - Lukas M. Simon
- Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Computational Biology, Neuherberg, Germany
| | - Hansjörg Schwertz
- Molecular Medicine Program
- Department of Internal Medicine
- Rocky Mountain Center for Occupational and Environmental Health
| | | | | | | | - Qing Li
- Huntsman Cancer Institute, Salt Lake City, Utah
| | | | | | | | | | | | | | - Paul F. Bray
- Molecular Medicine Program
- Department of Internal Medicine
| | | |
Collapse
|
86
|
Canault M, Alessi MC. RasGRP2 Structure, Function and Genetic Variants in Platelet Pathophysiology. Int J Mol Sci 2020; 21:E1075. [PMID: 32041177 PMCID: PMC7037602 DOI: 10.3390/ijms21031075] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 01/31/2020] [Accepted: 02/03/2020] [Indexed: 12/18/2022] Open
Abstract
RasGRP2 is calcium and diacylglycerol-regulated guanine nucleotide exchange factor I that activates Rap1, which is an essential signaling-knot in "inside-out" αIIbβ3 integrin activation in platelets. Inherited platelet function disorder caused by variants of RASGRP2 represents a new congenital bleeding disorder referred to as platelet-type bleeding disorder-18 (BDPLT18). We review here the structure of RasGRP2 and its functions in the pathophysiology of platelets and of the other cellular types that express it. We will also examine the different pathogenic variants reported so far as well as strategies for the diagnosis and management of patients with BDPLT18.
Collapse
Affiliation(s)
- Matthias Canault
- Aix Marseille University, INSERM, INRAE, C2VN, 13005 Marseille, France
| | - Marie-Christine Alessi
- Aix Marseille University, INSERM, INRAE, C2VN, 13005 Marseille, France
- Hematology laboratory, APHM, CHU Timone, 13005 Marseille, France
| |
Collapse
|
87
|
Tumor-educated platelet as liquid biopsy in lung cancer patients. Crit Rev Oncol Hematol 2020; 146:102863. [PMID: 31935617 DOI: 10.1016/j.critrevonc.2020.102863] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 12/28/2019] [Accepted: 01/01/2020] [Indexed: 02/06/2023] Open
Abstract
Lung cancer is the most frequent cancer for males and third most frequent cancer for females. Targeted therapy drugs based on molecular alterations, such as angiogenesis inhibitors, epidermal growth factor receptor (EGFR) inhibitors, and anaplastic lymphoma kinase (ALK) inhibitors are important part of treatment of NSCLC. However, the quality of the available tumor biopsy and/or cytology material is sometimes not adequate to perform the necessary molecular testing, which has prompted the search for alternatives. This review examines the use of tumor-educated platelet (TEP) as a liquid biopsy in lung cancer patients. The development of sensitive and accurate techniques have made it possible to detect the specific genetic alterations for which targeted therapies are already available. Liquid biopsy offers opportunities to detect resistance mechanisms at an early stage. To conclude, tumor-educated platelet has the potential to be used as liquid biopsy for a variety of clinical and investigational applications.
Collapse
|
88
|
Piacentini L, Werba JP, Bono E, Saccu C, Tremoli E, Spirito R, Colombo GI. Genome-Wide Expression Profiling Unveils Autoimmune Response Signatures in the Perivascular Adipose Tissue of Abdominal Aortic Aneurysm. Arterioscler Thromb Vasc Biol 2019; 39:237-249. [PMID: 30567485 DOI: 10.1161/atvbaha.118.311803] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective- Perivascular adipose tissue (PVAT) is thought to play a role in vascular homeostasis and in the pathogenesis of large vessel diseases, including abdominal aortic aneurysm (AAA). Herein, we tested the hypothesis that locally restricted transcriptional profiles characterize PVAT surrounding AAA, indicating specific dysfunctions associated with the disease. Approach and Results- Using a paired sample design to limit the effects of interindividual variation, we performed a microarray-based investigation of the PVAT transcriptome in 30 patients with AAA, comparing the adipose layer of the dilated abdominal aorta with that of the not-dilated aortic neck in each patient. Furthermore, we used a state-of-the-art data mining procedure to remove the effect of confounders produced by high-throughput gene expression techniques. We found substantial differences in PVAT gene expression clearly distinguishing the dilated from the not-dilated aorta, which increased in number and magnitude with increasing AAA diameter. Comparisons with other adipose depots (omental or subcutaneous fat) confirmed that gene expression changes are locally restricted. We dissected putative mechanisms associated with AAA PVAT dysfunction through a functional enrichment network analysis: both innate and adaptive immune-response genes along with genes related to cell-death pathways, metabolic processes of collagen, sphingolipids, aminoglycans, and extracellular matrix degradation were strongly overrepresented in PVAT of AAA compared with PVAT of the not-dilated aorta. Conclusions- Our results support a possible function of PVAT in AAA pathogenesis and suggest that AAA is an immunologic disease with an underlying autoimmune component. Interfering with these disease-specific pathways would clarify their precise role in AAA pathogenesis.
Collapse
Affiliation(s)
- Luca Piacentini
- From the Immunology and Functional Genomics Unit (L.P., E.B., G.I.C.), Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | - José Pablo Werba
- Atherosclerosis Prevention Unit (J.P.W.), Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | - Elisa Bono
- From the Immunology and Functional Genomics Unit (L.P., E.B., G.I.C.), Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | - Claudio Saccu
- Department of Cardiovascular Surgery of the University of Milan (C.S., R.S.), Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | - Elena Tremoli
- Scientific Direction (E.T.), Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | - Rita Spirito
- Department of Cardiovascular Surgery of the University of Milan (C.S., R.S.), Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | - Gualtiero Ivanoe Colombo
- From the Immunology and Functional Genomics Unit (L.P., E.B., G.I.C.), Centro Cardiologico Monzino, IRCCS, Milan, Italy
| |
Collapse
|
89
|
Washington AV, Esponda O, Gibson A. Platelet biology of the rapidly failing lung. Br J Haematol 2019; 188:641-651. [PMID: 31696941 DOI: 10.1111/bjh.16315] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 08/23/2019] [Accepted: 08/24/2019] [Indexed: 12/15/2022]
Abstract
Acute respiratory distress syndrome (ARDS) is characterized by a rapid-onset respiratory failure with a mortality rate of approximately 40%. This physiologic inflammatory process is mediated by disruption of the alveolar-vascular interface, leading to pulmonary oedema and impaired oxygen exchange, which often warrants mechanical ventilation to increase survival in the acute setting. One of the least understood aspects of ARDS is the role of the platelets in this process. Platelets, which protect vascular integrity, play a pivotal role in the progression and resolution of ARDS. The recent substantiation of the age-old theory that megakaryocytes are found in the lungs has rejuvenated interest in and raised new questions about the importance of platelets for pulmonary function. In addition to primary haemostasis, platelets provide a myriad of inflammatory functions that are poised to aid the innate immune system. This review focuses on the evidence for regulatory roles of platelets in pulmonary inflammation, with an emphasis on two receptors, CLEC-2 and TLT-1. Studies of these receptors identify novel pathways through which platelets may regulate vascular integrity and inflammation in the lungs, thereby influencing the development of ARDS.
Collapse
Affiliation(s)
- A Valance Washington
- Department of Biology, Molecular Science Research Center, University of Puerto Rico-Rio Piedras, San Juan, PR, USA
| | | | | |
Collapse
|
90
|
Arnason NA, Johannson F, Landrö R, Hardarsson B, Irsch J, Gudmundsson S, Rolfsson O, Sigurjonsson OE. Pathogen inactivation with amotosalen plus UVA illumination minimally impacts microRNA expression in platelets during storage under standard blood banking conditions. Transfusion 2019; 59:3727-3735. [PMID: 31674051 DOI: 10.1111/trf.15575] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 09/15/2019] [Accepted: 10/03/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND To reduce the risk of transfusion transmission infection, nucleic acid targeted methods have been developed to inactivate pathogens in PCs. miRNAs have been shown to play an important role in platelet function, and changes in the abundance of specific miRNAs during storage have been observed, as have perturbation effects related to pathogen inactivation (PI) methods. The aim of this work was to investigate the effects of PI on selected miRNAs during storage. STUDY DESIGN AND METHODS Using a pool and split strategy, 3 identical buffy coat PC units were generated from a pool of 24 whole blood donors. Each unit received a different treatment: 1) Untreated platelet control in platelet additive solution (C-PAS); 2) Amotosalen-UVA-treated platelets in PAS (PI-PAS); and 3) untreated platelets in donor plasma (U-PL). PCs were stored for 7 days under standard blood banking conditions. Standard platelet quality control (QC) parameters and 25 selected miRNAs were analyzed. RESULTS During the 7-day storage period, differences were found in several QC parameters relating to PI treatment and storage in plasma, but overall the three treatments were comparable. Out of 25 miRNA tested changes in regulation of 5 miRNA in PI-PAS and 3 miRNA U-PL where detected compared to C-PAS. A statistically significant difference was observed in down regulations miR-96-5p on Days 2 and 4, 61.9% and 61.8%, respectively, in the PI-PAS treatment. CONCLUSION Amotosalen-UVA treatment does not significantly alter the miRNA profile of platelet concentrates generated and stored using standard blood banking conditions.
Collapse
Affiliation(s)
- Niels Arni Arnason
- The Blood Bank, Landspitali - The National University Hospital of Iceland, Reykjavik, Iceland
| | - Freyr Johannson
- Department of Medicine, University of Iceland, Reykjavik, Iceland
| | - Ragna Landrö
- The Blood Bank, Landspitali - The National University Hospital of Iceland, Reykjavik, Iceland
| | - Björn Hardarsson
- The Blood Bank, Landspitali - The National University Hospital of Iceland, Reykjavik, Iceland
| | | | - Sveinn Gudmundsson
- The Blood Bank, Landspitali - The National University Hospital of Iceland, Reykjavik, Iceland
| | - Ottar Rolfsson
- Department of Medicine, University of Iceland, Reykjavik, Iceland
| | - Olafur E Sigurjonsson
- The Blood Bank, Landspitali - The National University Hospital of Iceland, Reykjavik, Iceland.,School of Science and Engineering, Reykjavik University, Reykjavik, Iceland
| |
Collapse
|
91
|
Bhatlekar S, Basak I, Edelstein LC, Campbell RA, Lindsey CR, Italiano JE, Weyrich AS, Rowley JW, Rondina MT, Sola-Visner M, Bray PF. Anti-apoptotic BCL2L2 increases megakaryocyte proplatelet formation in cultures of human cord blood. Haematologica 2019; 104:2075-2083. [PMID: 30733267 PMCID: PMC6886406 DOI: 10.3324/haematol.2018.204685] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 01/30/2019] [Indexed: 12/23/2022] Open
Abstract
Apoptosis is a recognized limitation to generating large numbers of megakaryocytes in culture. The genes responsible have been rigorously studied in vivo in mice, but are poorly characterized in human culture systems. As CD34-positive (+) cells isolated from human umbilical vein cord blood were differentiated into megakaryocytes in culture, two distinct cell populations were identified by flow cytometric forward and side scatter: larger size, lower granularity (LLG), and smaller size, higher granularity (SHG). The LLG cells were CD41aHigh CD42aHigh phosphatidylserineLow, had an electron microscopic morphology similar to mature bone marrow megakaryocytes, developed proplatelets, and displayed a signaling response to platelet agonists. The SHG cells were CD41aLowCD42aLowphosphatidylserineHigh, had a distinctly apoptotic morphology, were unable to develop proplatelets, and showed no signaling response. Screens of differentiating megakaryocytes for expression of 24 apoptosis genes identified BCL2L2 as a novel candidate megakaryocyte apoptosis regulator. Lentiviral BCL2L2 overexpression decreased megakaryocyte apoptosis, increased CD41a+ LLG cells, and increased proplatelet formation by 58%. An association study in 154 healthy donors identified a significant positive correlation between platelet number and platelet BCL2L2 mRNA levels. This finding was consistent with the observed increase in platelet-like particles derived from cultured megakaryocytes over-expressing BCL2L2 BCL2L2 also induced small, but significant increases in thrombin-induced platelet-like particle αIIbβ3 activation and P-selectin expression. Thus, BCL2L2 restrains apoptosis in cultured megakaryocytes, promotes proplatelet formation, and is associated with platelet number. BCL2L2 is a novel target for improving megakaryocyte and platelet yields in in vitro culture systems.
Collapse
Affiliation(s)
- Seema Bhatlekar
- Program in Molecular Medicine and Department of Internal Medicine, University of Utah, Salt Lake City, UT
| | - Indranil Basak
- Program in Molecular Medicine and Department of Internal Medicine, University of Utah, Salt Lake City, UT
| | - Leonard C Edelstein
- Cardeza Foundation for Hematologic Research, Thomas Jefferson University, Philadelphia, PA
| | - Robert A Campbell
- Program in Molecular Medicine and Department of Internal Medicine, University of Utah, Salt Lake City, UT
| | - Cory R Lindsey
- Cardeza Foundation for Hematologic Research, Thomas Jefferson University, Philadelphia, PA
| | | | - Andrew S Weyrich
- Program in Molecular Medicine and Department of Internal Medicine, University of Utah, Salt Lake City, UT
| | - Jesse W Rowley
- Program in Molecular Medicine and Department of Internal Medicine, University of Utah, Salt Lake City, UT
| | - Matthew T Rondina
- Program in Molecular Medicine and Department of Internal Medicine, University of Utah, Salt Lake City, UT
- George E. Wahlen VAMC GRECC, Salt Lake City, UT
| | | | - Paul F Bray
- Program in Molecular Medicine and Department of Internal Medicine, University of Utah, Salt Lake City, UT
- Division of Hematology and Hematologic Malignancies, Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
92
|
Guo L, Rondina MT. The Era of Thromboinflammation: Platelets Are Dynamic Sensors and Effector Cells During Infectious Diseases. Front Immunol 2019; 10:2204. [PMID: 31572400 PMCID: PMC6753373 DOI: 10.3389/fimmu.2019.02204] [Citation(s) in RCA: 154] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 08/30/2019] [Indexed: 12/12/2022] Open
Abstract
Platelets are anucleate cells produced by megakaryocytes. In recent years, a robust body of literature supports the evolving role of platelets as key sentinel and effector cells in infectious diseases, especially critical in bridging hemostatic, inflammatory, and immune continuums. Upon intravascular pathogen invasion, platelets can directly sense viral, parasitic, and bacterial infections through pattern recognition receptors and integrin receptors or pathogen: immunoglobulin complexes through Fc and complement receptors—although our understanding of these interactions remains incomplete. Constantly scanning for areas of injury or inflammation as they circulate in the vasculature, platelets also indirectly respond to pathogen invasion through interactions with leukocytes and the endothelium. Following antigen recognition, platelets often become activated. Through a diverse repertoire of mechanisms, activated platelets can directly sequester or kill pathogens, or facilitate pathogen clearance by activating macrophages and neutrophils, promoting neutrophil extracellular traps (NETs) formation, forming platelet aggregates and microthrombi. At times, however, platelet activation may also be injurious to the host, exacerbating inflammation and promoting endothelial damage and thrombosis. There are many gaps in our understandings of the role of platelets in infectious diseases. However, with the emergence of advanced technologies, our knowledge is increasing. In the current review, we mainly discuss these evolving roles of platelets under four different infectious pathogen infections, of which are dengue, malaria, Esterichia coli (E. coli) and staphylococcus aureus S. aureus, highlighting the complex interplay of these processes with hemostatic and thrombotic pathways.
Collapse
Affiliation(s)
- Li Guo
- University of Utah Molecular Medicine Program, Salt Lake City, UT, United States
| | - Matthew T Rondina
- University of Utah Molecular Medicine Program, Salt Lake City, UT, United States.,Department of Internal Medicine, University of Utah, Salt Lake City, UT, United States.,Department of Pathology, University of Utah, Salt Lake City, UT, United States.,George E. Wahlen VAMC Department of Internal Medicine and GRECC, Salt Lake City, UT, United States
| |
Collapse
|
93
|
Le Blanc J, Lordkipanidzé M. Platelet Function in Aging. Front Cardiovasc Med 2019; 6:109. [PMID: 31448291 PMCID: PMC6692461 DOI: 10.3389/fcvm.2019.00109] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 07/22/2019] [Indexed: 12/20/2022] Open
Abstract
Aging is associated with an increased incidence of cardiovascular disease and thrombosis. Platelets play a major role in maintaining hemostasis and in thrombus formation, making them a key player in thrombotic disorders. Whereas it is well-known that platelet aggregability is increased in vascular diseases, the contribution of age-related changes in platelet biology to cardiovascular risk is not well-understood. Several lines of evidence support that platelets from older subjects differ in their function and structure, making platelets more prone to activation and less sensitive to inhibition. These age-related changes could lead to platelet hyperactivity and to the development of a prothrombotic state in advanced age. This review will focus on platelet biochemical modifications during aging and on the mechanisms by which these alterations could lead to thrombotic disease.
Collapse
Affiliation(s)
- Jessica Le Blanc
- Research Center, Montreal Heart Institute, Montreal, QC, Canada.,Faculty of Pharmacy, Université de Montréal, Montreal, QC, Canada
| | - Marie Lordkipanidzé
- Research Center, Montreal Heart Institute, Montreal, QC, Canada.,Faculty of Pharmacy, Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
94
|
Madan N, Ghazi AR, Kong X, Chen ES, Shaw CA, Edelstein LC. Functionalization of CD36 cardiovascular disease and expression associated variants by interdisciplinary high throughput analysis. PLoS Genet 2019; 15:e1008287. [PMID: 31344026 PMCID: PMC6684090 DOI: 10.1371/journal.pgen.1008287] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 08/06/2019] [Accepted: 07/04/2019] [Indexed: 12/22/2022] Open
Abstract
CD36 is a platelet membrane glycoprotein whose engagement with oxidized low-density lipoprotein (oxLDL) results in platelet activation. The CD36 gene has been associated with platelet count, platelet volume, as well as lipid levels and CVD risk by genome-wide association studies. Platelet CD36 expression levels have been shown to be associated with both the platelet oxLDL response and an elevated risk of thrombo-embolism. Several genomic variants have been identified as associated with platelet CD36 levels, however none have been conclusively demonstrated to be causative. We screened 81 expression quantitative trait loci (eQTL) single nucleotide polymorphisms (SNPs) associated with platelet CD36 expression by a Massively Parallel Reporter Assay (MPRA) and analyzed the results with a novel Bayesian statistical method. Ten eQTLs located 13kb to 55kb upstream of the CD36 transcriptional start site of transcript ENST00000309881 and 49kb to 92kb upstream of transcript ENST00000447544, demonstrated significant transcription shifts between their minor and major allele in the MPRA assay. Of these, rs2366739 and rs1194196, separated by only 20bp, were confirmed by luciferase assay to alter transcriptional regulation. In addition, electromobility shift assays demonstrated differential DNA:protein complex formation between the two alleles of this locus. Furthermore, deletion of the genomic locus by CRISPR/Cas9 in K562 and Meg-01 cells results in upregulation of CD36 transcription. These data indicate that we have identified a variant that regulates expression of CD36, which in turn affects platelet function. To assess the clinical relevance of our findings we used the PhenoScanner tool, which aggregates large scale GWAS findings; the results reinforce the clinical relevance of our variants and the utility of the MPRA assay. The study demonstrates a generalizable paradigm for functional testing of genetic variants to inform mechanistic studies, support patient management and develop precision therapies. Platelets are anucleate cells that are best known as regulators of vascular hemostasis and thrombosis but also play important roles in cancer, angiogenesis, and inflammation. CD36 is a platelet surface marker that can activate platelet in response to oxidized low density lipoprotein (oxLDL). CD36 has been associated with numerous cardiovascular traits in human including blood lipid levels, platelet count, and cardiovascular disease prevalence in human genetic studies. Human variability in platelet CD36 levels are associated with the platelet response to oxLDL. However, the genetic mechanisms responsible for the variability of CD36 levels are unknown. We examined 81 genetic variants associated with CD36 levels for functionality using a high-throughput assay. Of the ten variants that were identified in that assay, one doublet, rs2366739 and rs1194196, were confirmed using additional molecular and cellular assays. Deletion of the genomic region containing rs2366739 and rs1194196 resulted in overexpression of CD36 in a cell culture system. This finding indicates a control locus which can serve as a potential target in modulating CD36 expression and altering platelet function in cardiovascular disease.
Collapse
Affiliation(s)
- Namrata Madan
- Cardeza Foundation for Hematologic Research/Department of Medicine, Sidney Kimmel Medical School, Thomas Jefferson University, Philadelphia, PA, United States of America
| | - Andrew R. Ghazi
- Department of Quantitative and Computational Biosciences, Baylor College of Medicine, Houston, TX, United States of America
| | - Xianguo Kong
- Cardeza Foundation for Hematologic Research/Department of Medicine, Sidney Kimmel Medical School, Thomas Jefferson University, Philadelphia, PA, United States of America
| | - Edward S. Chen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States of America
| | - Chad A. Shaw
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States of America
- Department of Statistics, Rice University, Houston, TX, United States of America
| | - Leonard C. Edelstein
- Cardeza Foundation for Hematologic Research/Department of Medicine, Sidney Kimmel Medical School, Thomas Jefferson University, Philadelphia, PA, United States of America
- * E-mail:
| |
Collapse
|
95
|
Espinosa-Parrilla Y, Gonzalez-Billault C, Fuentes E, Palomo I, Alarcón M. Decoding the Role of Platelets and Related MicroRNAs in Aging and Neurodegenerative Disorders. Front Aging Neurosci 2019; 11:151. [PMID: 31312134 PMCID: PMC6614495 DOI: 10.3389/fnagi.2019.00151] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 06/11/2019] [Indexed: 12/22/2022] Open
Abstract
Platelets are anucleate cells that circulate in blood and are essential components of the hemostatic system. During aging, platelet numbers decrease and their aggregation capacity is reduced. Platelet dysfunctions associated with aging can be linked to molecular alterations affecting several cellular systems that include cytoskeleton rearrangements, signal transduction, vesicular trafficking, and protein degradation. Age platelets may adopt a phenotype characterized by robust secretion of extracellular vesicles that could in turn account for about 70-90% of blood circulating vesicles. Interestingly these extracellular vesicles are loaded with messenger RNAs and microRNAs that may have a profound impact on protein physiology at the systems level. Age platelet dysfunction is also associated with accumulation of reactive oxygen species. Thereby understanding the mechanisms of aging in platelets as well as their age-dependent dysfunctions may be of interest when evaluating the contribution of aging to the onset of age-dependent pathologies, such as those affecting the nervous system. In this review we summarize the findings that link platelet dysfunctions to neurodegenerative diseases including Alzheimer's Disease, Parkinson's Disease, Multiple Sclerosis, Huntington's Disease, and Amyotrophic Lateral Sclerosis. We discuss the role of platelets as drivers of protein dysfunctions observed in these pathologies, their association with aging and the potential clinical significance of platelets, and related miRNAs, as peripheral biomarkers for diagnosis and prognosis of neurodegenerative diseases.
Collapse
Affiliation(s)
- Yolanda Espinosa-Parrilla
- School of Medicine, Universidad de Magallanes, Punta Arenas, Chile
- Laboratory of Molecular Medicine-LMM, Center for Education, Healthcare and Investigation-CADI, Universidad de Magallanes, Punta Arenas, Chile
- Thematic Task Force on Healthy Aging, CUECH Research Network, Santiago, Chile
| | - Christian Gonzalez-Billault
- Thematic Task Force on Healthy Aging, CUECH Research Network, Santiago, Chile
- Laboratory of Cell and Neuronal Dynamics, Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism GERO, Santiago, Chile
- The Buck Institute for Research on Aging, Novato, CA, United States
| | - Eduardo Fuentes
- Thematic Task Force on Healthy Aging, CUECH Research Network, Santiago, Chile
- Thrombosis Research Center, Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences and Research Center for Aging, Universidad de Talca, Talca, Chile
| | - Ivan Palomo
- Thematic Task Force on Healthy Aging, CUECH Research Network, Santiago, Chile
- Thrombosis Research Center, Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences and Research Center for Aging, Universidad de Talca, Talca, Chile
| | - Marcelo Alarcón
- Thematic Task Force on Healthy Aging, CUECH Research Network, Santiago, Chile
- Thrombosis Research Center, Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences and Research Center for Aging, Universidad de Talca, Talca, Chile
| |
Collapse
|
96
|
Barbati C, Stefanini L, Colasanti T, Cipriano E, Celia A, Gabriele G, Vomero M, Ceccarelli F, Spinelli FR, Finucci A, Speziali M, Orso G, Margiotta DPE, Conti F, Violi F, Afeltra A, Valesini G, Alessandri C. Anti-D4GDI antibodies activate platelets in vitro: a possible link with thrombocytopenia in primary antiphospholipid syndrome. Arthritis Res Ther 2019; 21:161. [PMID: 31262358 PMCID: PMC6604387 DOI: 10.1186/s13075-019-1947-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 06/17/2019] [Indexed: 11/17/2022] Open
Abstract
Background Thrombocytopenia is a manifestation associated with primary antiphospholipid syndrome (PAPS), and many studies have stressed the leading role played by platelets in the pathogenesis of antiphospholipid syndrome (APS). Platelets are highly specialized cells, and their activation involves a series of rapid rearrangements of the actin cytoskeleton. Recently, we described the presence of autoantibodies against D4GDI (Rho GDP dissociation inhibitor beta, ARHGDIB) in the serum of a large subset of SLE patients, and we observed that anti-D4GDI antibodies activated the cytoskeleton remodeling of lymphocytes by inhibiting D4GDI and allowing the upregulation of Rho GTPases, such as Rac1. Proteomic and transcriptomic studies indicate that D4GDI is very abundant in platelets, and small GTPases of the RHO family are critical regulators of actin dynamics in platelets. Methods We enrolled 38 PAPS patients, 15 patients carrying only antiphospholipid antibodies without clinical criteria of APS (aPL carriers) and 20 normal healthy subjects. Sera were stored at − 20 °C to perform an ELISA test to evaluate the presence of anti-D4GDI antibodies. Then, we purified autoantibodies anti-D4GDI from patient sera. These antibodies were used to conduct in vitro studies on platelet activation. Results We identified anti-D4GDI antibodies in sera from 18/38 (47%) patients with PAPS, in sera from 2/15(13%) aPL carriers, but in no sera from normal healthy subjects. Our in vitro results showed a significant 30% increase in the activation of integrin αIIbβ3 upon stimulation of platelets from healthy donors preincubated with the antibody anti-D4GDI purified from the serum of APS patients. Conclusions In conclusion, we show here that antibodies anti-D4GDI are present in the sera of PAPS patients and can prime platelet activation, explaining, at least in part, the pro-thrombotic state and the thrombocytopenia of PAPS patients. These findings may lead to improved diagnosis and treatment of APS.
Collapse
Affiliation(s)
- C Barbati
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Viale del Policlinico, 155, Rome, Italy.
| | - L Stefanini
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Viale del Policlinico, 155, Rome, Italy
| | - T Colasanti
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Viale del Policlinico, 155, Rome, Italy
| | - E Cipriano
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Viale del Policlinico, 155, Rome, Italy
| | - A Celia
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Viale del Policlinico, 155, Rome, Italy
| | - G Gabriele
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Viale del Policlinico, 155, Rome, Italy
| | - M Vomero
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Viale del Policlinico, 155, Rome, Italy
| | - F Ceccarelli
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Viale del Policlinico, 155, Rome, Italy
| | - F R Spinelli
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Viale del Policlinico, 155, Rome, Italy
| | - A Finucci
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Viale del Policlinico, 155, Rome, Italy
| | - M Speziali
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Viale del Policlinico, 155, Rome, Italy
| | - G Orso
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Viale del Policlinico, 155, Rome, Italy
| | - D P E Margiotta
- Department of Immuno-Rheumatology, Campus Bio-Medico, University of Rome, Rome, Italy
| | - F Conti
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Viale del Policlinico, 155, Rome, Italy
| | - F Violi
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Viale del Policlinico, 155, Rome, Italy
| | - A Afeltra
- Department of Immuno-Rheumatology, Campus Bio-Medico, University of Rome, Rome, Italy
| | - G Valesini
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Viale del Policlinico, 155, Rome, Italy
| | - C Alessandri
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Viale del Policlinico, 155, Rome, Italy
| |
Collapse
|
97
|
Best MG, In 't Veld SGJG, Sol N, Wurdinger T. RNA sequencing and swarm intelligence-enhanced classification algorithm development for blood-based disease diagnostics using spliced blood platelet RNA. Nat Protoc 2019; 14:1206-1234. [PMID: 30894694 DOI: 10.1038/s41596-019-0139-5] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 01/17/2019] [Indexed: 12/12/2022]
Abstract
Blood-based diagnostics tests, using individual or panels of biomarkers, may revolutionize disease diagnostics and enable minimally invasive therapy monitoring. However, selection of the most relevant biomarkers from liquid biosources remains an immense challenge. We recently presented the thromboSeq pipeline, which enables RNA sequencing and cancer classification via self-learning and swarm intelligence-enhanced bioinformatics algorithms using blood platelet RNA. Here, we provide the wet-lab protocol for the generation of platelet RNA-sequencing libraries and the dry-lab protocol for the development of swarm intelligence-enhanced machine-learning-based classification algorithms. The wet-lab protocol includes platelet RNA isolation, mRNA amplification, and preparation for next-generation sequencing. The dry-lab protocol describes the automated FASTQ file pre-processing to quantified gene counts, quality controls, data normalization and correction, and swarm intelligence-enhanced support vector machine (SVM) algorithm development. This protocol enables platelet RNA profiling from 500 pg of platelet RNA and allows automated and optimized biomarker panel selection. The wet-lab protocol can be performed in 5 d before sequencing, and the algorithm development can be completed in 2 d, depending on computational resources. The protocol requires basic molecular biology skills and a basic understanding of Linux and R. In all, with this protocol, we aim to enable the scientific community to test platelet RNA for diagnostic algorithm development.
Collapse
Affiliation(s)
- Myron G Best
- Department of Neurosurgery, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center, Amsterdam, the Netherlands. .,Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center, Amsterdam, the Netherlands. .,Brain Tumor Center Amsterdam, Amsterdam UMC, VU University Medical Center, Amsterdam, the Netherlands.
| | - Sjors G J G In 't Veld
- Department of Neurosurgery, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center, Amsterdam, the Netherlands.,Brain Tumor Center Amsterdam, Amsterdam UMC, VU University Medical Center, Amsterdam, the Netherlands
| | - Nik Sol
- Brain Tumor Center Amsterdam, Amsterdam UMC, VU University Medical Center, Amsterdam, the Netherlands.,Department of Neurology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center, Amsterdam, the Netherlands
| | - Thomas Wurdinger
- Department of Neurosurgery, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center, Amsterdam, the Netherlands. .,Brain Tumor Center Amsterdam, Amsterdam UMC, VU University Medical Center, Amsterdam, the Netherlands.
| |
Collapse
|
98
|
Basak I, Bhatlekar S, Manne B, Stoller M, Hugo S, Kong X, Ma L, Rondina MT, Weyrich AS, Edelstein LC, Bray PF. miR-15a-5p regulates expression of multiple proteins in the megakaryocyte GPVI signaling pathway. J Thromb Haemost 2019; 17:511-524. [PMID: 30632265 PMCID: PMC6397079 DOI: 10.1111/jth.14382] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Indexed: 12/22/2022]
Abstract
Essentials The action of microRNAs (miRs) in human megakaryocyte signaling is largely unknown. Cord blood-derived human megakaryocytes (MKs) were used to test the function of candidate miRs. miR-15a-5p negatively regulated MK GPVI-mediated αIIbβ3 activation and α-granule release. miR-15a-5p acts as a potential "master-miR" regulating genes in the MK GPVI signaling pathway. SUMMARY: Background Megakaryocytes (MKs) invest their progeny platelets with proteins and RNAs. MicroRNAs (miRs), which inhibit mRNA translation into protein, are abundantly expressed in MKs and platelets. Although platelet miRs have been associated with platelet reactivity and disease, there is a paucity of information on the function of miRs in human MKs. Objective To identify MK miRs that regulate the GPVI signaling pathway in the MK-platelet lineage. Methods Candidate miRs associated with GPVI-mediated platelet aggregation were tested for functionality in cultured MKs derived from cord blood. Results An unbiased, transcriptome-wide screen in 154 healthy donors identified platelet miR-15a-5p as significantly negatively associated with CRP-induced platelet aggregation. Platelet agonist dose-response curves demonstrated activation of αIIbβ3 in suspensions of cord blood-derived cultured MKs. Overexpression and knockdown of miR-15a-5p in these MKs reduced and enhanced, respectively, CRP-induced αIIbβ3 activation but did not alter thrombin or ADP stimulation. FYN, SRGN, FCER1G, MYLK. and PRKCQ, genes involved in GPVI signaling, were identified as miR-15a-5p targets and were inhibited or de-repressed in MKs with miR-15a-5p overexpression or inhibition, respectively. Lentiviral overexpression of miR-15a-5p also inhibited GPVI-FcRγ-mediated phosphorylation of Syk and PLCγ2, GPVI downstream signaling molecules, but effects of miR-15a-5p on αIIbβ3 activation did not extend to other ITAM-signaling receptors (FcγRIIa and CLEC-2). Conclusion Cord blood-derived MKs are a useful human system for studying the functional effects of candidate platelet genes. miR-15a-5p is a potential "master-miR" for specifically regulating GPVI-mediated MK-platelet signaling. Targeting miR-15a-5p may have therapeutic potential in hemostasis and thrombosis.
Collapse
Affiliation(s)
- I. Basak
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA; and Division of General Internal Medicine, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84112, USA; and George E. Wahlen VAMC, Salt Lake City, UT, 84148
| | - S. Bhatlekar
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA; and Division of General Internal Medicine, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84112, USA; and George E. Wahlen VAMC, Salt Lake City, UT, 84148
| | - B.K. Manne
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA; and Division of General Internal Medicine, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84112, USA; and George E. Wahlen VAMC, Salt Lake City, UT, 84148
| | - M. Stoller
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA; and Division of General Internal Medicine, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84112, USA; and George E. Wahlen VAMC, Salt Lake City, UT, 84148
| | - S. Hugo
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA; and Division of General Internal Medicine, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84112, USA; and George E. Wahlen VAMC, Salt Lake City, UT, 84148
| | - X. Kong
- The Cardeza Foundation for Hematologic Research and the Department of Medicine, Thomas Jefferson University, Jefferson Medical College, Philadelphia, PA 19107
| | - L. Ma
- The Cardeza Foundation for Hematologic Research and the Department of Medicine, Thomas Jefferson University, Jefferson Medical College, Philadelphia, PA 19107
| | - M. T. Rondina
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA; and Division of General Internal Medicine, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84112, USA; and George E. Wahlen VAMC, Salt Lake City, UT, 84148
| | - A. S. Weyrich
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA; and Division of General Internal Medicine, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84112, USA; and George E. Wahlen VAMC, Salt Lake City, UT, 84148
| | - L. C. Edelstein
- The Cardeza Foundation for Hematologic Research and the Department of Medicine, Thomas Jefferson University, Jefferson Medical College, Philadelphia, PA 19107
| | - P. F. Bray
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA; and Division of General Internal Medicine, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84112, USA; and George E. Wahlen VAMC, Salt Lake City, UT, 84148
- Division of Hematology and Hematologic Malignancies, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
99
|
Halushka PV, Goodwin AJ, Halushka MK. Opportunities for microRNAs in the Crowded Field of Cardiovascular Biomarkers. ANNUAL REVIEW OF PATHOLOGY 2019; 14:211-238. [PMID: 30332561 PMCID: PMC6442682 DOI: 10.1146/annurev-pathmechdis-012418-012827] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Cardiovascular diseases exist across all developed countries. Biomarkers that can predict or diagnose diseases early in their pathogeneses can reduce their morbidity and mortality in afflicted individuals. microRNAs are small regulatory RNAs that modulate translation and have been identified as potential fluid-based biomarkers across numerous maladies. We describe the current state of cardiovascular disease biomarkers across a range of diseases, including myocardial infarction, acute coronary syndrome, myocarditis, hypertension, heart failure, heart transplantation, aortic stenosis, diabetic cardiomyopathy, atrial fibrillation, and sepsis. We present the current understanding of microRNAs as possible biomarkers in these categories and where their best opportunities exist to enter clinical practice.
Collapse
Affiliation(s)
- Perry V Halushka
- Department of Pharmacology, South Carolina Clinical and Translational Research Institute, Medical University of South Carolina, Charleston, South Carolina 29425, USA;
- Department of Medicine, South Carolina Clinical and Translational Research Institute, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | - Andrew J Goodwin
- Division of Pulmonary, Critical Care, Allergy, and Sleep Medicine, Medical University of South Carolina, Charleston, South Carolina 29425, USA;
| | - Marc K Halushka
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA;
| |
Collapse
|
100
|
Heffron SP, Marier C, Parikh M, Fisher EA, Berger JS. Severe obesity and bariatric surgery alter the platelet mRNA profile. Platelets 2019; 30:967-974. [PMID: 30388921 PMCID: PMC6642854 DOI: 10.1080/09537104.2018.1536261] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Mechanisms explaining the relationship between obesity and cardiovascular disease (CVD) are needed. Despite growing recognition of the importance of the anucleate platelet transcriptome, low levels of RNA in platelets make assessment difficult. We sought to perform unbiased platelet RNA profiling in obesity by performing a prospective study of severe obesity and weight loss via bariatric surgery on platelet characteristics and mRNA profile in 26 pre-menopausal, non-diabetic women (31.6 ± 8.4 years; BMI 43.0 ± 6.5 kg/m2) who underwent sleeve gastrectomy. Totally, 10 women of similar age with normal BMI served as controls. Platelet activation via flow cytometry was assessed before and after surgery. RNA-sequencing (RNAseq) was performed on platelet isolates from a subset of 13 subjects (eight obese women and five normal-BMI subjects). Platelet count, size, and age did not differ between control and obese women. However, platelet surface P-selectin and CD40 were higher in obesity. RNAseq demonstrated 629 differentially abundant transcripts in obesity. Notably, S100A9 and AGER, established markers of cardiovascular risk, were two of the most highly upregulated transcripts (each > 2.5 fold). At 6 months post-operatively, subjects lost 26.1 ± 5.8% body weight and inducible platelet P-selectin expression was reduced. Expression of 170 transcripts was affected by surgery, but only a small fraction (46/629) were genes found altered in obesity. We demonstrate that obesity is associated with an altered platelet transcriptome and increased platelet activation, which is partly attenuated by bariatric surgery. These observations suggest that platelets may contribute to increased cardiovascular risk in obesity through a variety of mechanisms.
Collapse
Affiliation(s)
- Sean P Heffron
- Department of Medicine, Leon H. Charney Division of Cardiology and the Center for the Prevention of Cardiovascular Disease;,Address for Correspondence: Sean P. Heffron MD, MS, MSc, Instructor in Medicine, Leon H. Charney Division of Cardiology, New York University School of Medicine, 227 East 30th St., #834, New York, NY 10016, Tel: -1- 646-501-2735,
| | - Christian Marier
- Genome Technology Core Laboratory, New York University School of Medicine, New York, NY
| | - Manish Parikh
- Department of Surgery, New York University Langone Medical Center
| | - Edward A Fisher
- Department of Medicine, Leon H. Charney Division of Cardiology and the Center for the Prevention of Cardiovascular Disease
| | - Jeffrey S Berger
- Department of Medicine, Leon H. Charney Division of Cardiology and the Center for the Prevention of Cardiovascular Disease;,Department of Surgery, Division of Vascular Surgery, New York University Langone Medical Center
| |
Collapse
|