101
|
Rondina MT, Zimmerman GA. The Role of Platelets in Inflammation. Platelets 2019. [DOI: 10.1016/b978-0-12-813456-6.00028-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
|
102
|
|
103
|
Morales-Ortíz J, Deal V, Reyes F, Maldonado-Martínez G, Ledesma N, Staback F, Croft C, Pacheco A, Ortiz-Zuazaga H, Yost CC, Rowley JW, Madera B, John AS, Chen J, Lopez J, Rondina MT, Hunter R, Gibson A, Washington AV. Platelet-derived TLT-1 is a prognostic indicator in ALI/ARDS and prevents tissue damage in the lungs in a mouse model. Blood 2018; 132:2495-2505. [PMID: 30282800 PMCID: PMC6284217 DOI: 10.1182/blood-2018-03-841593] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 09/14/2018] [Indexed: 02/06/2023] Open
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) affect >200 000 individuals yearly with a 40% mortality rate. Although platelets are implicated in the progression of ALI/ARDS, their exact role remains undefined. Triggering receptor expressed in myeloid cells (TREM)-like transcript 1 (TLT-1) is found on platelets, binds fibrinogen, and mediates clot formation. We hypothesized that platelets use TLT-1 to manage the progression of ALI/ARDS. Here we retrospectively measure plasma levels of soluble TLT-1 (sTLT-1) from the ARDS Network clinical trial and show that patients whose sTLT-1 levels were >1200 pg/mL had nearly twice the mortality risk as those with <1200 pg/mL (P < .001). After correcting for confounding factors such as creatinine levels, Acute Physiology And Chronic Health Evaluation III scores, age, platelet counts, and ventilation volume, sTLT-1 remains significant, suggesting that sTLT-1 is an independent prognostic factor (P < .0001). These data point to a role for TLT-1 during the progression of ALI/ARDS. We use a murine lipopolysaccharide-induced ALI model and demonstrate increased alveolar bleeding, aberrant neutrophil transmigration and accumulation associated with decreased fibrinogen deposition, and increased pulmonary tissue damage in the absence of TLT-1. The loss of TLT-1 resulted in an increased proportion of platelet-neutrophil conjugates (43.73 ± 24.75% vs 8.92 ± 2.4% in wild-type mice), which correlated with increased neutrophil death. Infusion of sTLT-1 restores normal fibrinogen deposition and reduces pulmonary hemorrhage by 40% (P ≤ .001) and tissue damage by 25% (P ≤ .001) in vivo. Our findings suggest that TLT-1 uses fibrinogen to govern the transition between inflammation and hemostasis and facilitate controlled leukocyte transmigration during the progression of ARDS.
Collapse
Affiliation(s)
| | - Victoria Deal
- Division of Natural Sciences, Maryville College, Maryville, TN
| | - Fiorella Reyes
- Department of Biology, University of Puerto Rico-Rio Piedras, San Juan, Puerto Rico
| | | | - Nahomy Ledesma
- Department of Biology, University of Puerto Rico-Rio Piedras, San Juan, Puerto Rico
| | - Franklin Staback
- Department of Biology, University of Puerto Rico-Rio Piedras, San Juan, Puerto Rico
| | - Cheyanne Croft
- Division of Natural Sciences, Maryville College, Maryville, TN
| | - Amanda Pacheco
- Department of Biology, University of Puerto Rico-Rio Piedras, San Juan, Puerto Rico
| | - Humberto Ortiz-Zuazaga
- Department of Computer Science, University of Puerto Rico-Rio Piedras, San Juan, Puerto Rico
| | - C Christian Yost
- Department of Pediatrics/Neonatology and Molecular Medicine Program and
| | - Jesse W Rowley
- Department of Internal Medicine and Molecular Medicine Program, University of Utah School of Medicine, Salt Lake City, UT
| | - Bismark Madera
- Department of Biology, University of Puerto Rico-Rio Piedras, San Juan, Puerto Rico
| | - Alex St John
- Bloodworks Northwest Research Institute, Seattle, WA; and
| | - Junmei Chen
- Bloodworks Northwest Research Institute, Seattle, WA; and
| | - Jose Lopez
- Bloodworks Northwest Research Institute, Seattle, WA; and
| | - Matthew T Rondina
- Department of Internal Medicine and Molecular Medicine Program, University of Utah School of Medicine, Salt Lake City, UT
- Geriatric Research, Education and Clinical Center, Department of Medicine, George E. Wahlen VA Medical Center, Salt Lake City, UT
| | - Robert Hunter
- Retroviral Research Center, Universidad Central del Caribe, Bayamón, Puerto Rico
| | - Angelia Gibson
- Division of Natural Sciences, Maryville College, Maryville, TN
| | - A Valance Washington
- Department of Biology, University of Puerto Rico-Rio Piedras, San Juan, Puerto Rico
| |
Collapse
|
104
|
Fisher MH, Di Paola J. Genomics and transcriptomics of megakaryocytes and platelets: Implications for health and disease. Res Pract Thromb Haemost 2018; 2:630-639. [PMID: 30349880 PMCID: PMC6178711 DOI: 10.1002/rth2.12129] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 06/03/2018] [Indexed: 01/07/2023] Open
Abstract
The field of megakaryocyte and platelet biology has been transformed with the implementation of high throughput sequencing. The use of modern sequencing technologies has led to the discovery of causative mutations in congenital platelet disorders and has been a useful tool in uncovering many other mechanisms of altered platelet formation and function. Although the understanding of the presence of RNA in platelets is relatively novel, mRNA and miRNA expression profiles are being shown to play an increasingly important role in megakaryopoiesis and platelet function in normal physiology as well as in disease states. Understanding the genetic perturbations underlying platelet dysfunction provides insight into normal megakaryopoiesis and thrombopoiesis, as well as guiding the development of novel therapeutics.
Collapse
Affiliation(s)
- Marlie H. Fisher
- Department of PediatricsUniversity of Colorado School of MedicineAuroraColorado
- Medical Scientist Training ProgramUniversity of Colorado School of MedicineAuroraColorado
| | - Jorge Di Paola
- Department of PediatricsUniversity of Colorado School of MedicineAuroraColorado
- Medical Scientist Training ProgramUniversity of Colorado School of MedicineAuroraColorado
| |
Collapse
|
105
|
Tilburg J, Adili R, Nair TS, Hawley ME, Tuk DC, Jackson M, Spronk HM, Versteeg HH, Carey TE, van Vlijmen BJM, Maracle CX, Holinstat M. Characterization of hemostasis in mice lacking the novel thrombosis susceptibility gene Slc44a2. Thromb Res 2018; 171:155-159. [PMID: 30312801 DOI: 10.1016/j.thromres.2018.09.057] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 09/01/2018] [Accepted: 09/24/2018] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Recent genome wide association studies (GWAS) identified a novel susceptibility locus for thrombosis, harbouring the SLC44A2 gene which encodes the Solute Carrier Family 44 Member 2 protein (SLC44A2). Thus far, SLC44A2 has not been studied in the context of thrombosis, and may be a unique contributor to thrombotic disease. Here we utilize mice lacking SLC44A2 (Slc44a2-/-) to evaluate a possible role of SLC44A2 in hemostasis. METHODS Slc44a2-/- mice were evaluated in key aspects of normal hemostasis including a challenge of vascular damage by applying laser induced injury to the cremaster muscle arteriole. RESULTS Slc44a2-/- mice had comparable levels of thrombin generation and gene expression of coagulation related genes, as compared to littermate wild type controls. Lower levels of circulating plasma Von Willebrand factor (VWF) were measured in Slc44a2-/- mice, while no difference in VWF multimerization or vascular localization was detected. Upon in vivo laser injury of the cremaster arterioles, we detected an impairment of clot formation for Slc44a2-/- mice. CONCLUSIONS Although mice lacking SLC44A2 are normal for several hemostasis parameters, we do observe a reduction of plasma VWF levels and an altered response upon vascular damage, which suggests that SLC44A2 contributes to hemostasis upon injury. These findings are in line with the reported GWAS data and support further research on SLC44A2 in thrombosis.
Collapse
Affiliation(s)
- Julia Tilburg
- Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands; Department of Internal Medicine, Division of Thrombosis and Hemostasis, Leiden University Medical Center, Leiden, The Netherlands.
| | - Reheman Adili
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA
| | - Thankam S Nair
- Kresge Hearing Research Institute, Department of Otolaryngology/Head and Neck Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Megan E Hawley
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA
| | - David C Tuk
- Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands; Department of Internal Medicine, Division of Thrombosis and Hemostasis, Leiden University Medical Center, Leiden, The Netherlands
| | - Madeline Jackson
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA
| | - Henri M Spronk
- Laboratory for Clinical Thrombosis and Hemostasis, Department of Internal Medicine and Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Henri H Versteeg
- Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands; Department of Internal Medicine, Division of Thrombosis and Hemostasis, Leiden University Medical Center, Leiden, The Netherlands
| | - Thomas E Carey
- Kresge Hearing Research Institute, Department of Otolaryngology/Head and Neck Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Bart J M van Vlijmen
- Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands; Department of Internal Medicine, Division of Thrombosis and Hemostasis, Leiden University Medical Center, Leiden, The Netherlands
| | - Chrissta X Maracle
- Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands; Department of Internal Medicine, Division of Thrombosis and Hemostasis, Leiden University Medical Center, Leiden, The Netherlands.
| | - Michael Holinstat
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA; Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
106
|
Abstract
SIGNIFICANCE Platelets are anucleate blood cells that are involved in hemostasis and thrombosis. Although no longer able to generate ribonucleic acid (RNA) de novo, platelets contain messenger RNA (mRNA), YRNA fragments, and premature microRNAs (miRNAs) that they inherit from megakaryocytes. Recent Advances: Novel sequencing techniques have helped identify the unexpectedly large number of RNA species present in platelets. Throughout their life time, platelets can process the pre-existing pool of premature miRNA to give the fully functional miRNA that can regulate platelet protein expression and function. CRITICAL ISSUES Platelets make a major contribution to the circulating miRNA pool but platelet activation can have major consequences on Dicer levels and thus miRNA maturation, which has implications for studies that are focused on screening-stored platelets. FUTURE DIRECTIONS It will be important to determine the importance of platelets as donors for miRNA-containing microvesicles that can be taken up and processed by other (particularly vascular) cells, thus contributing to homeostasis as well as disease progression. Antioxid. Redox Signal. 29, 902-921.
Collapse
Affiliation(s)
- Amro Elgheznawy
- 1 Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University , Frankfurt am Main, Germany .,2 German Center for Cardiovascular Research (DZHK) , Partner site Rhein-Main, Frankfurt am Main, Germany
| | - Ingrid Fleming
- 1 Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University , Frankfurt am Main, Germany .,2 German Center for Cardiovascular Research (DZHK) , Partner site Rhein-Main, Frankfurt am Main, Germany
| |
Collapse
|
107
|
Platelet communication with the vascular wall: role of platelet-derived microparticles and non-coding RNAs. Clin Sci (Lond) 2018; 132:1875-1888. [PMID: 30185611 DOI: 10.1042/cs20180580] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 07/31/2018] [Accepted: 08/09/2018] [Indexed: 12/24/2022]
Abstract
Platelets play an important role in vascular homeostasis through their interaction with circulating blood cells as well as the vascular wall. Platelet-mediated communication with other cells can take the form of direct cell-cell interactions via membrane receptors or indirectly through the release of different soluble factors stored in their granules as well as through the release of microparticles. The latter carry different proteins and RNAs which are transferred to the target cells. The aim of this review is to discuss the role of platelet-derived factors, adhesion molecules as well as RNAs as mediators of the cross-talk between platelets and the vessel wall.
Collapse
|
108
|
Day DB, Clyde MA, Xiang J, Li F, Cui X, Mo J, Gong J, Weschler CJ, Zhang Y, Zhang JJ. Age modification of ozone associations with cardiovascular disease risk in adults: a potential role for soluble P-selectin and blood pressure. J Thorac Dis 2018; 10:4643-4652. [PMID: 30174917 DOI: 10.21037/jtd.2018.06.135] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background Studies have suggested that age increases susceptibility to ozone-associated mortality, but the underlying mechanisms are unclear. In a previous study, personal exposure to ozone was significantly associated with a platelet activation biomarker, plasma soluble P-selectin (sCD62P), and blood pressure in 89 healthy adults, aged 22-52 years. The present study examines whether age modifies these associations in the same adults and in additional adults. Methods Interaction terms of age and exposure were analyzed using hierarchical Bayesian mixed effects ridge regressions. Data from a similar additional study involving 71 healthy participants, aged 19-26 years, were pooled with the data from the first study to evaluate age effect modification when more young adults were added to the analysis. Results In the 89 adults, significant age interactions were observed for past 24-hour and 2-week ozone exposures and sCD62P. Based on the pooled data (89 plus 71 adults), a 10 ppb increase in 24-hour ozone exposure was associated with increases in sCD62P and systolic blood pressure (SBP) by 22.3% (95% CI: 14.3%, 31.2%) and 1.35 (-0.18, 2.84) mmHg, respectively, at age 25; these values increased to 48.6% (32.7%, 65.1%) and 4.98 (2.56, 7.35) mmHg, respectively, at age 40. Conclusions These results mechanistically suggest that increasing age enhances cardiovascular effects of ozone.
Collapse
Affiliation(s)
- Drew B Day
- Global Health Institute and Nicholas School of the Environment, Duke University, Durham, NC, USA
| | - Merlise A Clyde
- Department of Statistical Science, Duke University, Durham, NC, USA
| | - Jianbang Xiang
- Department of Building Science, Tsinghua University, Beijing 100084, China.,Beijing Key Laboratory of Indoor Air Quality Evaluation and Control, Beijing 100084, China
| | - Feng Li
- Department of Respiratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Xiaoxing Cui
- Global Health Institute and Nicholas School of the Environment, Duke University, Durham, NC, USA
| | - Jinhan Mo
- Department of Building Science, Tsinghua University, Beijing 100084, China.,Beijing Key Laboratory of Indoor Air Quality Evaluation and Control, Beijing 100084, China
| | - Jicheng Gong
- College of Environmental Sciences and Engineering and Beijing Innovation Center for Engineering Science and Advanced Technology, Peking University, Beijing 100871, China
| | - Charles J Weschler
- Department of Building Science, Tsinghua University, Beijing 100084, China.,Beijing Key Laboratory of Indoor Air Quality Evaluation and Control, Beijing 100084, China.,Environmental and Occupational Health Sciences Institute, Rutgers University, Robert Wood Johnson Medical School, Piscataway, NJ, USA
| | - Yinping Zhang
- Department of Building Science, Tsinghua University, Beijing 100084, China.,Beijing Key Laboratory of Indoor Air Quality Evaluation and Control, Beijing 100084, China
| | - Junfeng Jim Zhang
- Global Health Institute and Nicholas School of the Environment, Duke University, Durham, NC, USA.,College of Environmental Sciences and Engineering and Beijing Innovation Center for Engineering Science and Advanced Technology, Peking University, Beijing 100871, China.,Duke Kunshan University, Kunshan 215347, China
| |
Collapse
|
109
|
Functional redundancy between RAP1 isoforms in murine platelet production and function. Blood 2018; 132:1951-1962. [PMID: 30131434 DOI: 10.1182/blood-2018-03-838714] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 08/11/2018] [Indexed: 01/14/2023] Open
Abstract
RAP GTPases, important regulators of cellular adhesion, are abundant signaling molecules in the platelet/megakaryocytic lineage. However, mice lacking the predominant isoform, RAP1B, display a partial platelet integrin activation defect and have a normal platelet count, suggesting the existence of a RAP1-independent pathway to integrin activation in platelets and a negligible role for RAP GTPases in megakaryocyte biology. To determine the importance of individual RAP isoforms on platelet production and on platelet activation at sites of mechanical injury or vascular leakage, we generated mice with megakaryocyte-specific deletion (mKO) of Rap1a and/or Rap1b Interestingly, Rap1a/b-mKO mice displayed a marked macrothrombocytopenia due to impaired proplatelet formation by megakaryocytes. In platelets, RAP isoforms had redundant and isoform-specific functions. Deletion of RAP1B, but not RAP1A, significantly reduced α-granule secretion and activation of the cytoskeleton regulator RAC1. Both isoforms significantly contributed to thromboxane A2 generation and the inside-out activation of platelet integrins. Combined deficiency of RAP1A and RAP1B markedly impaired platelet aggregation, spreading, and clot retraction. Consistently, thrombus formation in physiological flow conditions was abolished in Rap1a/b-mKO, but not Rap1a-mKO or Rap1b-mKO, platelets. Rap1a/b-mKO mice were strongly protected from experimental thrombosis and exhibited a severe defect in hemostasis after mechanical injury. Surprisingly, Rap1a/b-mKO platelets were indistinguishable from controls in their ability to prevent blood-lymphatic mixing during development and hemorrhage at sites of inflammation. In summary, our studies demonstrate an essential role for RAP1 signaling in platelet integrin activation and a critical role in platelet production. Although important for hemostatic/thrombotic plug formation, platelet RAP1 signaling is dispensable for vascular integrity during development and inflammation.
Collapse
|
110
|
Jessica MO, Fiorella R, Ocatavio S, Linnette R, Nahomy L, Kanth MB, Bismarck M, Rondina MT, Valance WA. TLT-1-CONTROLS EARLY THROMBUS FORMATION AND STABILITY BY FACILITATING AIIBB3 OUTSIDE-IN SIGNALING IN MICE. ACTA ACUST UNITED AC 2018; 6:1143-1149. [PMID: 30931337 DOI: 10.21474/ijar01/7469] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Platelets regulate inflammation as well as hemostasis. Inflammatory insults often induce hemostatic function through mechanisms that are not always understood. The triggering receptor expressed in myeloid cells (TREM)-like transcript 1 (TLT-1) is an abundantly expressed platelet receptor and its deletion leads to hemorrhage and edema after lipopolysaccharide and TNF-α treatment. To define a role for TLT-1 in immune derived bleeding we used a CXCL-2 mediated local inflammatory reaction in the vessels of the cremaster muscle of treml1 -/- and wild type mice. Our whole mount immunofluorescent staining of the cremaster muscle demonstrated a 50% reduction in clot size and increased extravasation of plasma molecules in treml1 -/- mice compared to wild type. We demonstrate that the decreased clotting in treml1 -/- mice is associated with a 2X reduction in integrin β3 phosphorylation on residue Y773 after platelet activation, which is consistent with treml1 -/- mice displaying reduced outside-in signaling and smaller thrombi. We further substantiate TLT-1's role in the regulation of immune derived bleeding using the reverse arthus reaction and demonstrate TLT-1's role in thrombosis using the thromboplastin initiated and collagen/epinephrine models of pulmonary embolism. Thus, the data presented here demonstrate that TLT-1 regulates early clot formation though the stabilization of αIIbβ3 outside-in signaling.
Collapse
Affiliation(s)
| | - Reyes Fiorella
- Laboratory of Anatomy and Cell Biology, Universidad Central del Caribe, Bayamón PR
| | - Santiago Ocatavio
- Laboratory of Anatomy and Cell Biology, Universidad Central del Caribe, Bayamón PR
| | - Rivera Linnette
- Laboratory of Anatomy and Cell Biology, Universidad Central del Caribe, Bayamón PR
| | - Ledesma Nahomy
- University of Puerto Rico-Rio Piedras, Department of Biology
| | - Manne B Kanth
- Molecular Medicine Program and Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, Utah
| | - Madera Bismarck
- University of Puerto Rico-Rio Piedras, Department of Biology
| | - Matthew T Rondina
- Molecular Medicine Program and Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, Utah.,Department of Medicine and the George E. Whalen VAMC GRECC; Salt Lake City, Utah
| | - Washington A Valance
- University of Puerto Rico-Rio Piedras, Department of Biology.,Laboratory of Anatomy and Cell Biology, Universidad Central del Caribe, Bayamón PR
| |
Collapse
|
111
|
Quantification of microRNA levels in plasma - Impact of preanalytical and analytical conditions. PLoS One 2018; 13:e0201069. [PMID: 30024941 PMCID: PMC6053236 DOI: 10.1371/journal.pone.0201069] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 07/06/2018] [Indexed: 02/07/2023] Open
Abstract
Numerous studies have reported a potential role for circulating microRNAs as biomarkers in a wide variety of diseases. However, there is a critical reproducibility challenge some of which might be due to differences in preanalytical and/or analytical factors. Thus, in the current study we systematically investigated the impact of selected preanalytical and analytical variables on the measured microRNA levels in plasma. Similar levels of microRNA were found in platelet-poor plasma obtained by dual compared to prolonged single centrifugation. In contrast, poor correlation was observed between measurements in standard plasma compared to platelet-poor plasma. The correlation between quantitative real-time PCR and droplet digital PCR was found to be good, contrary to TaqMan Low Density Array and single TaqMan assays where no correlation could be demonstrated. Dependent on the specific microRNA measured and the normalization strategy used, the intra- and inter-assay variation of quantitative real-time PCR were found to be 4.2–6.8% and 10.5–31.4%, respectively. Using droplet digital PCR the intra-assay variation was 4.4–20.1%, and the inter-assay variation 5.7–26.7%. Plasma preparation and microRNA purification were found to account for 39–73% of the total intra-assay variation, dependent on the microRNA measured and the normalization strategy used. In conclusion, our study highlighted the importance of reporting comprehensive methodological information when publishing, allowing others to perform validation studies where preanalytical and analytical variables as causes for divergent results can be minimized. Furthermore, if microRNAs are to become routinely used diagnostic or prognostic biomarkers, the differences in plasma microRNA levels between health and diseased subjects must exceed the high preanalytical and analytical variability.
Collapse
|
112
|
Stefanini L, Bergmeier W. RAP GTPases and platelet integrin signaling. Platelets 2018; 30:41-47. [PMID: 29863951 PMCID: PMC6312509 DOI: 10.1080/09537104.2018.1476681] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 04/26/2018] [Accepted: 04/27/2018] [Indexed: 12/31/2022]
Abstract
Platelets are highly specialized cells that continuously patrol the vasculature to ensure its integrity (hemostasis). At sites of vascular injury, they are able to respond to trace amounts of agonists and to rapidly transition from an anti-adhesive/patrolling to an adhesive state (integrin inside-out activation) required for hemostatic plug formation. Pathological conditions that disturb the balance in the underlying signaling processes can lead to unwanted platelet activation (thrombosis) or to an increased bleeding risk. The small GTPases of the RAP subfamily, highly expressed in platelets, are critical regulators of cell adhesion, cytoskeleton remodeling, and MAP kinase signaling. Studies by our group and others demonstrate that RAP GTPases, in particular RAP1A and RAP1B, are the key molecular switches that turn on platelet activation/adhesiveness at sites of injury. In this review, we will summarize major findings on the role of RAP GTPases in platelet biology with a focus on the signaling pathways leading to the conversion of integrins to a high-affinity state.
Collapse
Affiliation(s)
- Lucia Stefanini
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - Wolfgang Bergmeier
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill (NC), USA
- McAllister Heart Institute, University of North Carolina at Chapel Hill (NC), USA
| |
Collapse
|
113
|
Taghizadeh M, Ahmadizad S, Naderi M. Effects of endurance training on hsa-miR-223, P2RY12 receptor expression and platelet function in type 2 diabetic patients. Clin Hemorheol Microcirc 2018. [PMID: 29526844 DOI: 10.3233/ch-170300] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Mahmoudreza Taghizadeh
- Department of Biological Sciences in Sport, Faculty of Sports Sciences and Health, Shahid Beheshti University, Iran
| | - Sajad Ahmadizad
- Department of Biological Sciences in Sport, Faculty of Sports Sciences and Health, Shahid Beheshti University, Iran
| | - Mahmood Naderi
- Cell-Based Therapies Research Center, Digestive Disease Research Institute, Tehran University of Medical Science, Tehran, Iran
| |
Collapse
|
114
|
Abstract
Platelets are equipped with RNA processing machineries, such as pre-mRNA splicing, pre-miRNA processing, and mRNA translation. Since platelets are devoid of a nucleus, most RNA transcripts in platelets are derived from megakaryocytes during thrombocytogenesis. However, platelets can also ingest RNA molecules during circulation and/or interaction with other cell types. Since platelets were first described by Bizzozero in 1881, their well-established role in hemostasis and thrombosis has been intensively studied. However, in the past decades, the list of biological processes in which platelets play an important role keeps expanding. In this review, we discuss how platelet RNA biomarker signatures can be altered in the presence of cancer.
Collapse
Affiliation(s)
- Nik Sol
- Department of Neurology, VU University Medical Center, Amsterdam, The Netherlands. .,Brain Tumor Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands.
| | - Thomas Wurdinger
- Brain Tumor Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands.,Department of Neurosurgery, VU University Medical Center, Amsterdam, The Netherlands.,Department of Neurology, Massachusetts General Hospital and Neuroscience Program, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
115
|
Miao X, Rahman MFU, Jiang L, Min Y, Tan S, Xie H, Lee L, Wang M, Malmström RE, Lui WO, Li N. Thrombin-reduced miR-27b attenuates platelet angiogenic activities in vitro via enhancing platelet synthesis of anti-angiogenic thrombospondin-1. J Thromb Haemost 2018; 16:791-801. [PMID: 29442415 DOI: 10.1111/jth.13978] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Indexed: 12/29/2022]
Abstract
Essentials It is unclear if platelet micro-RNAs can regulate de novo protein synthesis of platelets. Platelet de novo protein synthesis of thrombospondin-1 (TSP-1) was induced by thrombin. Thrombin stimulation in vitro altered platelet microRNA profiles, including decreased miR-27b. Decreased miR-27b hampers platelet angiogenic activities via enhancing de novo TSP-1 synthesis. SUMMARY Background Platelets can synthesize proteins upon activation. Platelets contain a number of microRNAs (miRNA) and a fully functional miRNA effector machinery. It is, however, unclear if platelet miRNAs can regulate protein synthesis of platelets, and whether the regulation may produce a physiological impact. Objectives To investigate if and how platelet miRNAs regulate de novo syntheses of angiogenic regulators and subsequently modulate platelet angiogenic activities. Methods and Results Microarray-based miRNA profiling showed that thrombin stimulation in vitro down- or up-regulated a number of platelet miRNAs, both in the total platelet miRNAs and in Ago2-associated miRNAs. Among those altered miRNAs, miR-27b was down-regulated in both the total and Ago2-immunoprecipitated miRNA profiles of platelets, which was confirmed by reverse transcription-quantitative PCR (RT-qPCR). Using western blotting assays, we showed that thrombin induced platelet de novo synthesis of thrombospondin-1, and that the level of thrombospondin-1 synthesis could reach a level of 3-5-fold higher than that before thrombin stimulation. With either the platelet precursor megakaryocyte cell line MEG-01 cells or mature platelets, we demonstrated that transfection of miR-27b mimic, but not the negative control of miRNA mimic, markedly reduced thrombospondin-1 protein levels. The latter subsequently enhanced platelet-dependent endothelial tube formation on matrigel. Conclusions Thrombin stimulation in vitro reduces platelet miR-27b levels that may markedly enhance thrombin-evoked platelet de novo synthesis of thrombospondin-1. Elevation of platelet miR-27b by transfection inhibits thrombospondin-1 synthesis, and subsequently enhances platelet pro-angiogenic activities. Hence, platelet activation-dependent reduction of miR-27b levels may represent a novel negative regulatory mechanism of platelet angiogenic activities.
Collapse
Affiliation(s)
- X Miao
- Department of Medicine-Solna, Clinical Pharmacology Group, Karolinska Institutet, Stockholm, Sweden
| | - M F-U Rahman
- Department of Medicine-Solna, Clinical Pharmacology Group, Karolinska Institutet, Stockholm, Sweden
| | - L Jiang
- Department of Medicine-Solna, Clinical Pharmacology Group, Karolinska Institutet, Stockholm, Sweden
- Department of Pathology, Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, Ningbo, China
| | - Y Min
- Department of Medicine-Solna, Clinical Pharmacology Group, Karolinska Institutet, Stockholm, Sweden
| | - S Tan
- Department of Medicine-Solna, Clinical Pharmacology Group, Karolinska Institutet, Stockholm, Sweden
| | - H Xie
- Department of Oncology-Pathology and Cancer Center Karolinska, Karolinska University Hospital-Solna, Stockholm, Sweden
- School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - L Lee
- Department of Oncology-Pathology and Cancer Center Karolinska, Karolinska University Hospital-Solna, Stockholm, Sweden
| | - M 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, China
| | - R E Malmström
- Department of Medicine-Solna, Clinical Pharmacology Group, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Pharmacology, Karolinska University Hospital-Solna, Stockholm, Sweden
| | - W-O Lui
- Department of Oncology-Pathology and Cancer Center Karolinska, Karolinska University Hospital-Solna, Stockholm, Sweden
| | - N Li
- Department of Medicine-Solna, Clinical Pharmacology Group, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Pharmacology, Karolinska University Hospital-Solna, Stockholm, Sweden
| |
Collapse
|
116
|
Kabiri Rad H, Mazaheri M, Dehghani Firozabadi A. Relative Expression of PBMC MicroRNA-133a Analysis in Patients Receiving Warfarin After Mechanical Heart Valve Replacement. Avicenna J Med Biotechnol 2018; 10:29-33. [PMID: 29296264 PMCID: PMC5742651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND MicroRNAs (miRNAs) are implicated in various biological processes including anticoagulation. However, the modulation of miRNA by pharmacological intervention such as warfarin treatment in patients receiving warfarin has not been disclosed yet. The aim of this study work was to assess the effect of warfarin drug on expression level of mir-133a-3p in patients with mechanical heart valve replacement. METHODS In this research, the expression level of miRNA-133a-3p was analyzed in Peripheral Blood Mononuclear Cells (PBMCs) from mechanical valve replacement patients who had received warfarin for at least 3 months continuously. Quantitative RT-PCR method was used for this assay. RESULTS Our findings indicated a significant diffrence between the rate of miR-133a-3p expression in individuals receiving warfarin and the control group (p<0.01). There was also a statistically significant difference in miR-133a-3p expression in patients with different ages (p<0.05) suggesting that the rate of miR-133a-3p expression in persons receiving warfarin is related to age. However, other variables like warfarin dose, International Normalized Ratio (INR), gender, and Body Mass Index (BMI) were not significantly effective on the miR-133a-3p experssion rate in individuals receving warfarin. CONCLUSION Based on our results, it can be concluded that miR-133a-3p is involved in the coagulation pathway. The recent result indicates that warfarin affects the expression of miR-133a. This expression may be potentially important for treatment by anticoagulants. Awareness of the time course of miRNA expression profile can improve efficiency of response to warfarin.
Collapse
Affiliation(s)
- Hamid Kabiri Rad
- International Campus, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mahta Mazaheri
- Department of Medical Genetics, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran,Corresponding author: Mahta Mazaheri, M.D., Ph.D., Department of Medical Genetics, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran, Tel: +98 035 37240171, E-mail:
| | - Ali Dehghani Firozabadi
- Yazd Cardiovascular Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
117
|
Pordzik J, Pisarz K, De Rosa S, Jones AD, Eyileten C, Indolfi C, Malek L, Postula M. The Potential Role of Platelet-Related microRNAs in the Development of Cardiovascular Events in High-Risk Populations, Including Diabetic Patients: A Review. Front Endocrinol (Lausanne) 2018; 9:74. [PMID: 29615970 PMCID: PMC5869202 DOI: 10.3389/fendo.2018.00074] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Accepted: 02/19/2018] [Indexed: 12/13/2022] Open
Abstract
Platelet activation plays a pivotal role in the development and progression of atherosclerosis, which often leads to potentially fatal ischemic events at later stages of the disease. Platelets and platelet microvesicles (PMVs) contain large amounts of microRNA (miRNA), which contributes largely to the pool of circulating miRNAs. Hence, they represent a promising option for the development of innovative diagnostic biomarkers, that can be specific for the underlying etiology. Circulating miRNAs can be responsible for intracellular communication and may have a biological effect on target cells. As miRNAs associated to both cardiovascular diseases (CVD) and diabetes mellitus can be measured by means of a wide array of techniques, they can be exploited as an innovative class of smart disease biomarkers. In this manuscript, we provide an outline of miRNAs associated with platelet function and reactivity (miR-223, miR-126, miR-197, miR-191, miR-21, miR-150, miR-155, miR-140, miR-96, miR-98) that should be evaluated as novel biomarkers to improve diagnostics and treatment of CVD.
Collapse
Affiliation(s)
- Justyna Pordzik
- Center for Preclinical Research and Technology CEPT, Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Warsaw, Poland
| | - Katarzyna Pisarz
- Center for Preclinical Research and Technology CEPT, Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Warsaw, Poland
| | - Salvatore De Rosa
- Division of Cardiology, Department of Medical and Surgical Sciences, “Magna Graecia” University, Catanzaro, Italy
| | - Axel Dyve Jones
- Center for Preclinical Research and Technology CEPT, Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Warsaw, Poland
| | - Ceren Eyileten
- Center for Preclinical Research and Technology CEPT, Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Warsaw, Poland
| | - Ciro Indolfi
- Division of Cardiology, Department of Medical and Surgical Sciences, “Magna Graecia” University, Catanzaro, Italy
- URT-CNR, Department of Medicine, Consiglio Nazionale delle Ricerche of IFC, Catanzaro, Italy
| | - Lukasz Malek
- Faculty of Rehabilitation, University of Physical Education, Warsaw, Poland
| | - Marek Postula
- Center for Preclinical Research and Technology CEPT, Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Warsaw, Poland
- *Correspondence: Marek Postula,
| |
Collapse
|
118
|
Müller K, Chatterjee M, Rath D, Geisler T. Platelets, inflammation and anti-inflammatory effects of antiplatelet drugs in ACS and CAD. Thromb Haemost 2017. [DOI: 10.1160/th14-11-0947] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
SummaryPlatelets play a pivotal role in chronic inflammation leading to progression of atherosclerosis and acute coronary events. Recent discoveries on novel mechanisms and platelet-dependent inflammatory targets underpin the role of platelets to maintain a chronic inflammatory condition in cardiovascular disease. There is strong and clinically relevant crosslink between chronic inflammation and platelet activation. Antiplatelet therapy is a cornerstone in the prevention and treatment of acute cardiovascular events. The benefit of antiplatelet agents has mainly been attributed to their direct anti-aggregatory impact. Some anti-inflammatory off-target effects have also been described. However, it is unclear whether these effects are secondary due to inhibition of platelet activation or are caused by direct distinct mechanisms interfering with inflammatory pathways. This article will highlight novel platelet associated targets that contribute to inflammation in cardiovascular disease and elucidate mechanisms by which currently available antiplatelet agents evolve anti-inflammatory capacities, in particular by carving out the differential mechanisms directly or indirectly affecting platelet mediated inflammation. It will further illustrate the prognostic impact of antiplatelet therapies by reducing inflammatory marker release in recent cardiovascular trials.
Collapse
|
119
|
Campbell RA, Franks Z, Bhatnagar A, Rowley JW, Manne BK, Supiano MA, Schwertz H, Weyrich AS, Rondina MT. Granzyme A in Human Platelets Regulates the Synthesis of Proinflammatory Cytokines by Monocytes in Aging. THE JOURNAL OF IMMUNOLOGY 2017; 200:295-304. [PMID: 29167233 DOI: 10.4049/jimmunol.1700885] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 10/30/2017] [Indexed: 12/13/2022]
Abstract
Dysregulated inflammation is implicated in the pathobiology of aging, yet platelet-leukocyte interactions and downstream cytokine synthesis in aging remains poorly understood. Platelets and monocytes were isolated from healthy younger (age <45, n = 37) and older (age ≥65, n = 30) adults and incubated together under autologous and nonautologous conditions. Synthesis of inflammatory cytokines by monocytes, alone or in the presence of platelets, was examined. Next-generation RNA-sequencing allowed for unbiased profiling of the platelet transcriptome in aging. Basal IL-8 and MCP-1 synthesis by monocytes alone did not differ between older and younger adults. However, in the presence of autologous platelets, monocytes from older adults synthesized greater IL-8 (41 ± 5 versus 9 ± 2 ng/ml, p < 0.0001) and MCP-1 (867 ± 150 versus 216 ± 36 ng/ml, p < 0.0001) than younger adults. Platelets from older adults were sufficient for upregulating the synthesis of inflammatory cytokines by monocytes. Using RNA-sequencing of platelets followed by validation via RT-PCR and immunoblot, we discovered that granzyme A (GrmA), a serine protease not previously identified in human platelets, increases with aging (∼9-fold versus younger adults, p < 0.05) and governs increased IL-8 and MCP-1 synthesis through TLR4 and caspase-1. Inhibiting GrmA reduced excessive IL-8 and MCP-1 synthesis in aging to levels similar to younger adults. In summary, human aging is associated with changes in the platelet transcriptome and proteome. GrmA is present and bioactive in human platelets, is higher in older adults, and controls the synthesis of inflammatory cytokines by monocytes. Alterations in the platelet molecular signature and signaling to monocytes may contribute to dysregulated inflammatory syndromes in older adults.
Collapse
Affiliation(s)
- Robert A Campbell
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112.,Division of General Internal Medicine, Department of Internal Medicine, School of Medicine, University of Utah, Salt Lake City, UT 84132
| | - Zechariah Franks
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112
| | - Anish Bhatnagar
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112
| | - Jesse W Rowley
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112.,Division of Respiratory, Critical Care, and Occupational Pulmonary Medicine, University of Utah, Salt Lake City, UT 84132
| | - Bhanu K Manne
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112
| | - Mark A Supiano
- George E. Wahlen Veterans Affairs Medical Center, Geriatric Research, Education and Clinical Center, Salt Lake City, UT 84148.,Division of Geriatrics, School of Medicine, University of Utah, Salt Lake City, UT 84132; and
| | - Hansjorg Schwertz
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112.,Division of Vascular Surgery, School of Medicine, University of Utah, Salt Lake City, UT 84132
| | - Andrew S Weyrich
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112.,Division of Respiratory, Critical Care, and Occupational Pulmonary Medicine, University of Utah, Salt Lake City, UT 84132
| | - Matthew T Rondina
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112; .,Division of General Internal Medicine, Department of Internal Medicine, School of Medicine, University of Utah, Salt Lake City, UT 84132.,George E. Wahlen Veterans Affairs Medical Center, Geriatric Research, Education and Clinical Center, Salt Lake City, UT 84148
| |
Collapse
|
120
|
Beaulieu L, Tanriverdi K, Freedman J, Clancy L. The role of RNA uptake in platelet heterogeneity. Thromb Haemost 2017; 117:948-961. [DOI: 10.1160/th16-11-0873] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 02/10/2017] [Indexed: 12/29/2022]
Abstract
SummaryThe role of platelets in regulating vascular homeostasis has expanded beyond mediation of haemostasis and thrombosis. The discovery of platelet RNA and the presence of subpopulations of platelets containing varying amounts of RNA suggest a role for platelet transcripts in vascular function. As the RNA in anucleated platelets is biologically functional and may transfer to other vascular cells, we hypothesised that platelet RNA diminishes over the lifespan of the platelet with diminishing platelet size due to horizontal cellular transfer. The purpose of this study is to determine if platelet RNA variance is the result of horizontal cellular transfer between platelets and other vascular cells. Utilising platelet sorting and RNA sequencing, we found that smaller platelets contained a more diverse set of transcripts than larger platelets. Further investigation using fluorescence imaging, gene expression analyses and in vitro and in vivo modelling revealed that platelets take up RNA from other vascular cells in a complex manner, revealing a dynamic role for platelets in modulating vascular homeostasis through bidirectional RNA transfer. The resultant RNA profile heterogeneity suggests unique functional roles for platelets dependent on size and complexity. This study expands our basic understanding of platelet function and heterogeneity and is the first to evaluate endogenous vascular RNA uptake and its relation to platelet processes. Our findings describe a novel endogenous phenomenon that can help elucidate the platelet’s role in these non-thrombotic and haemostatic fields, as well as present potential for diagnostic and therapeutic development.Supplementary Material to this article is available online at www.thrombosis-online.com.
Collapse
|
121
|
Friede K, Voora D. The need for sex-specific precision biomarkers for antiplatelet therapies. Future Cardiol 2017; 13:419-422. [PMID: 28832186 DOI: 10.2217/fca-2017-0043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Kevin Friede
- Department of Medicine, Duke University, Durham, NC 27710, USA
| | - Deepak Voora
- Department of Medicine, Duke University, Durham, NC 27710, USA.,Duke Center for Applied Genomics & Precision Medicine, Duke University, Durham, NC 27708, USA
| |
Collapse
|
122
|
Bavykin AS. Circulating microRNAs in the identification of biological fluids: A new approach to standardization of expression-based diagnostics. Mol Biol 2017. [DOI: 10.1134/s0026893317040045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
123
|
Platelet populations and priming in hematological diseases. Blood Rev 2017; 31:389-399. [PMID: 28756877 DOI: 10.1016/j.blre.2017.07.004] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 06/26/2017] [Accepted: 07/18/2017] [Indexed: 01/01/2023]
Abstract
In healthy subjects and patients with hematological diseases, platelet populations can be distinguished with different response spectra in hemostatic and vascular processes. These populations partly overlap, and are less distinct than those of leukocytes. The platelet heterogeneity is linked to structural properties, and is enforced by inequalities in the environment. Contributing factors are variability between megakaryocytes, platelet ageing, and positive or negative priming of platelets during their time in circulation. Within a hemostatic plug or thrombus, platelet heterogeneity is enhanced by unequal exposure to agonists, with populations of contracted platelets in the thrombus core, discoid platelets at the thrombus surface, patches of ballooned and procoagulant platelets forming thrombin, and coated platelets binding fibrin. Several pathophysiological hematological conditions can positively or negatively prime the responsiveness of platelet populations. As a consequence, in vivo and in vitro markers of platelet activation can differ in thrombotic and hematological disorders.
Collapse
|
124
|
Best MG, Vancura A, Wurdinger T. Platelet RNA as a circulating biomarker trove for cancer diagnostics. J Thromb Haemost 2017; 15:1295-1306. [PMID: 28671345 DOI: 10.1111/jth.13720] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Platelets are multifunctional cell fragments, circulating in blood in high abundance. Platelets assist in thrombus formation, sensing of pathogens entering the blood stream, signaling to immune cells, releasing vascular remodeling factors, and, negatively, enhancing cancer metastasis. Platelets are 'educated' by their environment, including in patients with cancer. Cancer cells appear to initiate intraplatelet signaling, resulting in splicing of platelet pre-mRNAs, and enhance secretion of cytokines. Platelets can induce leukocyte and endothelial cell modeling factors, for example, through adenine nucleotides (ATP), thereby facilitating extravasation of cancer cells. Besides releasing factors, platelets can also sequester RNAs and proteins released by cancer cells. Thus, platelets actively respond to queues from local and systemic conditions, thereby altering their transcriptome and molecular content. Platelets contain a rich repertoire of RNA species, including mRNAs, small non-coding RNAs and circular RNAs; although studies regarding the functionality of the various platelet RNA species require more attention. Recent advances in high-throughput characterization of platelet mRNAs revealed 10 to > 1000 altered mRNAs in platelets in the presence of disease. Hence, platelet RNA appears to be dynamically affected by pathological conditions, thus possibly providing opportunities to use platelet RNA as diagnostic, prognostic, predictive, or monitoring biomarkers. In this review, we cover the literature regarding the platelet RNA families, processing of platelet RNAs, and the potential application of platelet RNA as disease biomarkers.
Collapse
Affiliation(s)
- M G Best
- Department of Neurosurgery, VU University Medical Center, Amsterdam, the Netherlands
- Department of Pathology, VU University Medical Center, Amsterdam, the Netherlands
- Brain Tumor Center Amsterdam, VU University Medical Center, Amsterdam, the Netherlands
| | - A Vancura
- Department of Neurosurgery, VU University Medical Center, Amsterdam, the Netherlands
- Brain Tumor Center Amsterdam, VU University Medical Center, Amsterdam, the Netherlands
| | - T Wurdinger
- Department of Neurosurgery, VU University Medical Center, Amsterdam, the Netherlands
- Brain Tumor Center Amsterdam, VU University Medical Center, Amsterdam, the Netherlands
- Department of Neurology, Massachusetts General Hospital and Neuroscience Program, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
125
|
Fidler TP, Middleton EA, Rowley JW, Boudreau LH, Campbell RA, Souvenir R, Funari T, Tessandier N, Boilard E, Weyrich AS, Abel ED. Glucose Transporter 3 Potentiates Degranulation and Is Required for Platelet Activation. Arterioscler Thromb Vasc Biol 2017; 37:1628-1639. [PMID: 28663252 DOI: 10.1161/atvbaha.117.309184] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 06/13/2017] [Indexed: 11/16/2022]
Abstract
OBJECTIVE On activation, platelets increase glucose uptake, glycolysis, and glucose oxidation and consume stored glycogen. This correlation between glucose metabolism and platelet function is not well understood and even less is known about the role of glucose metabolism on platelet function in vivo. For glucose to enter a cell, it must be transported through glucose transporters. Here we evaluate the contribution of GLUT3 (glucose transporter 3) to platelet function to better understand glucose metabolism in platelets. APPROACH AND RESULTS Platelet-specific knockout of GLUT3 was generated by crossing mice harboring GLUT3 floxed allele to a PF4 (platelet factor 4)-driven Cre recombinase. In platelets, GLUT3 is localized primarily on α-granule membranes and under basal conditions facilitates glucose uptake into α-granules to be used for glycolysis. After activation, platelets degranulate and GLUT3 translocates to the plasma membrane, which is responsible for activation-mediated increased glucose uptake. In vivo, loss of GLUT3 in platelets increased survival in a collagen/epinephrine model of pulmonary embolism, and in a K/BxN model of autoimmune inflammatory disease, platelet-specific GLUT3 knockout mice display decreased disease progression. Mechanistically, loss of GLUT3 decreased platelet degranulation, spreading, and clot retraction. Decreased α-granule degranulation is due in part to an impaired ability of GLUT3 to potentiate exocytosis. CONCLUSIONS GLUT3-mediated glucose utilization and glycogenolysis in platelets promotes α-granule release, platelet activation, and postactivation functions.
Collapse
Affiliation(s)
- Trevor P Fidler
- From the Department of Pharmacology and Toxicology (T.P.F.), and Program in Molecular Medicine (T.P.F., E.A.M., J.W.R., R.A.C., A.S.W., E.D.A.), University of Utah, Salt Lake City; Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Carver College of Medicine, University of Iowa, Iowa City (T.P.F., R.S., T.F., E.D.A.); and Department of Infectious Diseases and Immunity, Centre de Recherche du Centre Hospitalier Universitaire de Québec and Faculté de Médecine de l'Université Laval, Quebec City, Canada (L.H.B., N.T., E.B.)
| | - Elizabeth A Middleton
- From the Department of Pharmacology and Toxicology (T.P.F.), and Program in Molecular Medicine (T.P.F., E.A.M., J.W.R., R.A.C., A.S.W., E.D.A.), University of Utah, Salt Lake City; Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Carver College of Medicine, University of Iowa, Iowa City (T.P.F., R.S., T.F., E.D.A.); and Department of Infectious Diseases and Immunity, Centre de Recherche du Centre Hospitalier Universitaire de Québec and Faculté de Médecine de l'Université Laval, Quebec City, Canada (L.H.B., N.T., E.B.)
| | - Jesse W Rowley
- From the Department of Pharmacology and Toxicology (T.P.F.), and Program in Molecular Medicine (T.P.F., E.A.M., J.W.R., R.A.C., A.S.W., E.D.A.), University of Utah, Salt Lake City; Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Carver College of Medicine, University of Iowa, Iowa City (T.P.F., R.S., T.F., E.D.A.); and Department of Infectious Diseases and Immunity, Centre de Recherche du Centre Hospitalier Universitaire de Québec and Faculté de Médecine de l'Université Laval, Quebec City, Canada (L.H.B., N.T., E.B.)
| | - Luc H Boudreau
- From the Department of Pharmacology and Toxicology (T.P.F.), and Program in Molecular Medicine (T.P.F., E.A.M., J.W.R., R.A.C., A.S.W., E.D.A.), University of Utah, Salt Lake City; Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Carver College of Medicine, University of Iowa, Iowa City (T.P.F., R.S., T.F., E.D.A.); and Department of Infectious Diseases and Immunity, Centre de Recherche du Centre Hospitalier Universitaire de Québec and Faculté de Médecine de l'Université Laval, Quebec City, Canada (L.H.B., N.T., E.B.)
| | - Robert A Campbell
- From the Department of Pharmacology and Toxicology (T.P.F.), and Program in Molecular Medicine (T.P.F., E.A.M., J.W.R., R.A.C., A.S.W., E.D.A.), University of Utah, Salt Lake City; Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Carver College of Medicine, University of Iowa, Iowa City (T.P.F., R.S., T.F., E.D.A.); and Department of Infectious Diseases and Immunity, Centre de Recherche du Centre Hospitalier Universitaire de Québec and Faculté de Médecine de l'Université Laval, Quebec City, Canada (L.H.B., N.T., E.B.)
| | - Rhonda Souvenir
- From the Department of Pharmacology and Toxicology (T.P.F.), and Program in Molecular Medicine (T.P.F., E.A.M., J.W.R., R.A.C., A.S.W., E.D.A.), University of Utah, Salt Lake City; Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Carver College of Medicine, University of Iowa, Iowa City (T.P.F., R.S., T.F., E.D.A.); and Department of Infectious Diseases and Immunity, Centre de Recherche du Centre Hospitalier Universitaire de Québec and Faculté de Médecine de l'Université Laval, Quebec City, Canada (L.H.B., N.T., E.B.)
| | - Trevor Funari
- From the Department of Pharmacology and Toxicology (T.P.F.), and Program in Molecular Medicine (T.P.F., E.A.M., J.W.R., R.A.C., A.S.W., E.D.A.), University of Utah, Salt Lake City; Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Carver College of Medicine, University of Iowa, Iowa City (T.P.F., R.S., T.F., E.D.A.); and Department of Infectious Diseases and Immunity, Centre de Recherche du Centre Hospitalier Universitaire de Québec and Faculté de Médecine de l'Université Laval, Quebec City, Canada (L.H.B., N.T., E.B.)
| | - Nicolas Tessandier
- From the Department of Pharmacology and Toxicology (T.P.F.), and Program in Molecular Medicine (T.P.F., E.A.M., J.W.R., R.A.C., A.S.W., E.D.A.), University of Utah, Salt Lake City; Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Carver College of Medicine, University of Iowa, Iowa City (T.P.F., R.S., T.F., E.D.A.); and Department of Infectious Diseases and Immunity, Centre de Recherche du Centre Hospitalier Universitaire de Québec and Faculté de Médecine de l'Université Laval, Quebec City, Canada (L.H.B., N.T., E.B.)
| | - Eric Boilard
- From the Department of Pharmacology and Toxicology (T.P.F.), and Program in Molecular Medicine (T.P.F., E.A.M., J.W.R., R.A.C., A.S.W., E.D.A.), University of Utah, Salt Lake City; Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Carver College of Medicine, University of Iowa, Iowa City (T.P.F., R.S., T.F., E.D.A.); and Department of Infectious Diseases and Immunity, Centre de Recherche du Centre Hospitalier Universitaire de Québec and Faculté de Médecine de l'Université Laval, Quebec City, Canada (L.H.B., N.T., E.B.)
| | - Andrew S Weyrich
- From the Department of Pharmacology and Toxicology (T.P.F.), and Program in Molecular Medicine (T.P.F., E.A.M., J.W.R., R.A.C., A.S.W., E.D.A.), University of Utah, Salt Lake City; Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Carver College of Medicine, University of Iowa, Iowa City (T.P.F., R.S., T.F., E.D.A.); and Department of Infectious Diseases and Immunity, Centre de Recherche du Centre Hospitalier Universitaire de Québec and Faculté de Médecine de l'Université Laval, Quebec City, Canada (L.H.B., N.T., E.B.)
| | - E Dale Abel
- From the Department of Pharmacology and Toxicology (T.P.F.), and Program in Molecular Medicine (T.P.F., E.A.M., J.W.R., R.A.C., A.S.W., E.D.A.), University of Utah, Salt Lake City; Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Carver College of Medicine, University of Iowa, Iowa City (T.P.F., R.S., T.F., E.D.A.); and Department of Infectious Diseases and Immunity, Centre de Recherche du Centre Hospitalier Universitaire de Québec and Faculté de Médecine de l'Université Laval, Quebec City, Canada (L.H.B., N.T., E.B.).
| |
Collapse
|
126
|
SNP in human ARHGEF3 promoter is associated with DNase hypersensitivity, transcript level and platelet function, and Arhgef3 KO mice have increased mean platelet volume. PLoS One 2017; 12:e0178095. [PMID: 28542600 PMCID: PMC5441597 DOI: 10.1371/journal.pone.0178095] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Accepted: 05/07/2017] [Indexed: 12/20/2022] Open
Abstract
Genome-wide association studies have identified a genetic variant at 3p14.3 (SNP rs1354034) that strongly associates with platelet number and mean platelet volume in humans. While originally proposed to be intronic, analysis of mRNA expression in primary human hematopoietic subpopulations reveals that this SNP is located directly upstream of the predominantly expressed ARHGEF3 isoform in megakaryocytes (MK). We found that ARHGEF3, which encodes a Rho guanine exchange factor, is dramatically upregulated during both human and murine MK maturation. We show that the SNP (rs1354034) is located in a DNase I hypersensitive region in human MKs and is an expression quantitative locus (eQTL) associated with ARHGEF3 expression level in human platelets, suggesting that it may be the causal SNP that accounts for the variations observed in human platelet traits and ARHGEF3 expression. In vitro human platelet activation assays revealed that rs1354034 is highly correlated with human platelet activation by ADP. In order to test whether ARHGEF3 plays a role in MK development and/or platelet function, we developed an Arhgef3 KO/LacZ reporter mouse model. Reflecting changes in gene expression, LacZ expression increases during MK maturation in these mice. Although Arhgef3 KO mice have significantly larger platelets, loss of Arhgef3 does not affect baseline MK or platelets nor does it affect platelet function or platelet recovery in response to antibody-mediated platelet depletion compared to littermate controls. In summary, our data suggest that modulation of ARHGEF3 gene expression in humans with a promoter-localized SNP plays a role in human MKs and human platelet function—a finding resulting from the biological follow-up of human genetic studies. Arhgef3 KO mice partially recapitulate the human phenotype.
Collapse
|
127
|
Sunderland N, Skroblin P, Barwari T, Huntley RP, Lu R, Joshi A, Lovering RC, Mayr M. MicroRNA Biomarkers and Platelet Reactivity: The Clot Thickens. Circ Res 2017; 120:418-435. [PMID: 28104774 DOI: 10.1161/circresaha.116.309303] [Citation(s) in RCA: 148] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 12/20/2016] [Accepted: 12/20/2016] [Indexed: 12/16/2022]
Abstract
Over the last few years, several groups have evaluated the potential of microRNAs (miRNAs) as biomarkers for cardiometabolic disease. In this review, we discuss the emerging literature on the role of miRNAs and other small noncoding RNAs in platelets and in the circulation, and the potential use of miRNAs as biomarkers for platelet activation. Platelets are a major source of miRNAs, YRNAs, and circular RNAs. By harnessing multiomics approaches, we may gain valuable insights into their potential function. Because not all miRNAs are detectable in the circulation, we also created a gene ontology annotation for circulating miRNAs using the gene ontology term extracellular space as part of blood plasma. Finally, we share key insights for measuring circulating miRNAs. We propose ways to standardize miRNA measurements, in particular by using platelet-poor plasma to avoid confounding caused by residual platelets in plasma or by adding RNase inhibitors to serum to reduce degradation. This should enhance comparability of miRNA measurements across different cohorts. We provide recommendations for future miRNA biomarker studies, emphasizing the need for accurate interpretation within a biological and methodological context.
Collapse
Affiliation(s)
- Nicholas Sunderland
- From the King's British Heart Foundation Centre, King's College London, United Kingdom (N.S., P.S., T.B., R.L., A.J., M.M.); and Centre for Cardiovascular Genetics, Institute of Cardiovascular Science, University College London, United Kingdom (R.P.H., R.C.L.)
| | - Philipp Skroblin
- From the King's British Heart Foundation Centre, King's College London, United Kingdom (N.S., P.S., T.B., R.L., A.J., M.M.); and Centre for Cardiovascular Genetics, Institute of Cardiovascular Science, University College London, United Kingdom (R.P.H., R.C.L.)
| | - Temo Barwari
- From the King's British Heart Foundation Centre, King's College London, United Kingdom (N.S., P.S., T.B., R.L., A.J., M.M.); and Centre for Cardiovascular Genetics, Institute of Cardiovascular Science, University College London, United Kingdom (R.P.H., R.C.L.)
| | - Rachael P Huntley
- From the King's British Heart Foundation Centre, King's College London, United Kingdom (N.S., P.S., T.B., R.L., A.J., M.M.); and Centre for Cardiovascular Genetics, Institute of Cardiovascular Science, University College London, United Kingdom (R.P.H., R.C.L.)
| | - Ruifang Lu
- From the King's British Heart Foundation Centre, King's College London, United Kingdom (N.S., P.S., T.B., R.L., A.J., M.M.); and Centre for Cardiovascular Genetics, Institute of Cardiovascular Science, University College London, United Kingdom (R.P.H., R.C.L.)
| | - Abhishek Joshi
- From the King's British Heart Foundation Centre, King's College London, United Kingdom (N.S., P.S., T.B., R.L., A.J., M.M.); and Centre for Cardiovascular Genetics, Institute of Cardiovascular Science, University College London, United Kingdom (R.P.H., R.C.L.)
| | - Ruth C Lovering
- From the King's British Heart Foundation Centre, King's College London, United Kingdom (N.S., P.S., T.B., R.L., A.J., M.M.); and Centre for Cardiovascular Genetics, Institute of Cardiovascular Science, University College London, United Kingdom (R.P.H., R.C.L.)
| | - Manuel Mayr
- From the King's British Heart Foundation Centre, King's College London, United Kingdom (N.S., P.S., T.B., R.L., A.J., M.M.); and Centre for Cardiovascular Genetics, Institute of Cardiovascular Science, University College London, United Kingdom (R.P.H., R.C.L.).
| |
Collapse
|
128
|
Abstract
The systems analysis of thrombosis seeks to quantitatively predict blood function in a given vascular wall and hemodynamic context. Relevant to both venous and arterial thrombosis, a Blood Systems Biology approach should provide metrics for rate and molecular mechanisms of clot growth, thrombotic risk, pharmacological response, and utility of new therapeutic targets. As a rapidly created multicellular aggregate with a polymerized fibrin matrix, blood clots result from hundreds of unique reactions within and around platelets propagating in space and time under hemodynamic conditions. Coronary artery thrombosis is dominated by atherosclerotic plaque rupture, complex pulsatile flows through stenotic regions producing high wall shear stresses, and plaque-derived tissue factor driving thrombin production. In contrast, venous thrombosis is dominated by stasis or depressed flows, endothelial inflammation, white blood cell-derived tissue factor, and ample red blood cell incorporation. By imaging vessels, patient-specific assessment using computational fluid dynamics provides an estimate of local hemodynamics and fractional flow reserve. High-dimensional ex vivo phenotyping of platelet and coagulation can now power multiscale computer simulations at the subcellular to cellular to whole vessel scale of heart attacks or strokes. In addition, an integrated systems biology approach can rank safety and efficacy metrics of various pharmacological interventions or clinical trial designs.
Collapse
Affiliation(s)
- Scott L Diamond
- From the Department of Chemical Engineering, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia.
| |
Collapse
|
129
|
Mills EW, Green R, Ingolia NT. Slowed decay of mRNAs enhances platelet specific translation. Blood 2017; 129:e38-e48. [PMID: 28213379 PMCID: PMC5409447 DOI: 10.1182/blood-2016-08-736108] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 12/31/2016] [Indexed: 11/20/2022] Open
Abstract
Platelets are anucleate cytoplasmic fragments that lack genomic DNA, but continue to synthesize protein using a pool of messenger RNAs (mRNAs), ribosomes, and regulatory small RNAs inherited from the precursor megakaryocyte (MK). The regulatory processes that shape the platelet transcriptome and the full scope of platelet translation have remained elusive. Using RNA sequencing (RNA-Seq) and ribosome profiling of primary human platelets, we show the platelet transcriptome encompasses a subset of transcripts detected by RNA-Seq analysis of in vitro-derived MK cells and that these platelet-enriched transcripts are broadly occupied by ribosomes. We use RNA-Seq of synchronized populations of in vitro-derived platelet-like particles to show that mRNA decay strongly shapes the nascent platelet transcriptome. Our data suggest that the decay of platelet mRNAs is slowed by the natural loss of the mRNA surveillance and ribosome rescue factor Pelota.
Collapse
Affiliation(s)
- Eric W Mills
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Embryology, Carnegie Institution of Washington, Baltimore, MD; and
| | - Rachel Green
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Nicholas T Ingolia
- Department of Embryology, Carnegie Institution of Washington, Baltimore, MD; and
- Department of Molecular Cell Biology, Center for RNA Systems Biology, Glenn Center for Aging Research, University of California Berkeley, Berkeley, CA
| |
Collapse
|
130
|
Kong X, Simon LM, Holinstat M, Shaw CA, Bray PF, Edelstein LC. Identification of a functional genetic variant driving racially dimorphic platelet gene expression of the thrombin receptor regulator, PCTP. Thromb Haemost 2017; 117:962-970. [PMID: 28251237 DOI: 10.1160/th16-09-0692] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 02/12/2017] [Indexed: 01/08/2023]
Abstract
Platelet activation in response to stimulation of the Protease Activated Receptor 4 (PAR4) receptor differs by race. One factor that contributes to this difference is the expression level of Phosphatidylcholine Transfer Protein (PCTP), a regulator of platelet PAR4 function. We have conducted an expression Quantitative Trait Locus (eQTL) analysis that identifies single nucleotide polymorphisms (SNPs) linked to the expression level of platelet genes. This analysis revealed 26 SNPs associated with the expression level of PCTP at genome-wide significance (p < 5×10-8). Using annotation from ENCODE and other public data we prioritised one of these SNPs, rs2912553, for functional testing. The allelic frequency of rs2912553 is racially-dimorphic, in concordance with the racially differential expression of PCTP. Reporter gene assays confirmed that the single nucleotide change caused by rs2912553 altered the transcriptional potency of the surrounding genomic locus. Electromobility shift assays, luciferase assays, and overexpression studies indicated a role for the megakaryocytic transcription factor GATA1. In summary, we have integrated multi-omic data to identify and functionalise an eQTL. This, along with the previously described relationship between PCTP and PAR4 function, allows us to characterise a genotype-phenotype relationship through the mechanism of gene expression.
Collapse
Affiliation(s)
| | | | | | | | | | - Leonard C Edelstein
- Leonard C. Edelstein, Department of Medicine Sidney Kimmel Medical College, Thomas Jefferson University, 1020 Locust Street, Suite 394, Philadelphia, PA 19107, USA, Tel.: +1 215 955 1797, Fax: +1 215 955 9170,
| |
Collapse
|
131
|
Joehanes R, Zhang X, Huan T, Yao C, Ying SX, Nguyen QT, Demirkale CY, Feolo ML, Sharopova NR, Sturcke A, Schäffer AA, Heard-Costa N, Chen H, Liu PC, Wang R, Woodhouse KA, Tanriverdi K, Freedman JE, Raghavachari N, Dupuis J, Johnson AD, O'Donnell CJ, Levy D, Munson PJ. Integrated genome-wide analysis of expression quantitative trait loci aids interpretation of genomic association studies. Genome Biol 2017; 18:16. [PMID: 28122634 PMCID: PMC5264466 DOI: 10.1186/s13059-016-1142-6] [Citation(s) in RCA: 128] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 12/20/2016] [Indexed: 12/21/2022] Open
Abstract
Background Identification of single nucleotide polymorphisms (SNPs) associated with gene expression levels, known as expression quantitative trait loci (eQTLs), may improve understanding of the functional role of phenotype-associated SNPs in genome-wide association studies (GWAS). The small sample sizes of some previous eQTL studies have limited their statistical power. We conducted an eQTL investigation of microarray-based gene and exon expression levels in whole blood in a cohort of 5257 individuals, exceeding the single cohort size of previous studies by more than a factor of 2. Results We detected over 19,000 independent lead cis-eQTLs and over 6000 independent lead trans-eQTLs, targeting over 10,000 gene targets (eGenes), with a false discovery rate (FDR) < 5%. Of previously published significant GWAS SNPs, 48% are identified to be significant eQTLs in our study. Some trans-eQTLs point toward novel mechanistic explanations for the association of the SNP with the GWAS-related phenotype. We also identify 59 distinct blocks or clusters of trans-eQTLs, each targeting the expression of sets of six to 229 distinct trans-eGenes. Ten of these sets of target genes are significantly enriched for microRNA targets (FDR < 5%). Many of these clusters are associated in GWAS with multiple phenotypes. Conclusions These findings provide insights into the molecular regulatory patterns involved in human physiology and pathophysiology. We illustrate the value of our eQTL database in the context of a recent GWAS meta-analysis of coronary artery disease and provide a list of targeted eGenes for 21 of 58 GWAS loci. Electronic supplementary material The online version of this article (doi:10.1186/s13059-016-1142-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Roby Joehanes
- The Framingham Heart Study and the Division of Intramural Research, National Heart, Lung and Blood Institute, National Institutes of Health, 73 Mt. Wayte Avenue, Suite 2, Framingham, MA, 01702, USA.,Mathematical and Statistical Computing Laboratory, Center for Information Technology, National Institutes of Health, Bethesda, MD, USA.,Institute for Aging Research, Hebrew SeniorLife, Harvard Medical School, Boston, MA, USA
| | - Xiaoling Zhang
- The Framingham Heart Study and the Division of Intramural Research, National Heart, Lung and Blood Institute, National Institutes of Health, 73 Mt. Wayte Avenue, Suite 2, Framingham, MA, 01702, USA.,Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA.,Section of Biomedical Genetics, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Tianxiao Huan
- The Framingham Heart Study and the Division of Intramural Research, National Heart, Lung and Blood Institute, National Institutes of Health, 73 Mt. Wayte Avenue, Suite 2, Framingham, MA, 01702, USA
| | - Chen Yao
- The Framingham Heart Study and the Division of Intramural Research, National Heart, Lung and Blood Institute, National Institutes of Health, 73 Mt. Wayte Avenue, Suite 2, Framingham, MA, 01702, USA
| | - Sai-Xia Ying
- Mathematical and Statistical Computing Laboratory, Center for Information Technology, National Institutes of Health, Bethesda, MD, USA
| | - Quang Tri Nguyen
- Mathematical and Statistical Computing Laboratory, Center for Information Technology, National Institutes of Health, Bethesda, MD, USA
| | - Cumhur Yusuf Demirkale
- Mathematical and Statistical Computing Laboratory, Center for Information Technology, National Institutes of Health, Bethesda, MD, USA
| | - Michael L Feolo
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, USA
| | - Nataliya R Sharopova
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, USA
| | - Anne Sturcke
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, USA
| | - Alejandro A Schäffer
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, USA
| | - Nancy Heard-Costa
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Han Chen
- School of Public Health, Harvard University, Boston, MA, USA.,Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
| | - Po-Ching Liu
- DNA Sequencing and Genomics Core, National Institutes of Health, Bethesda, MD, USA
| | - Richard Wang
- DNA Sequencing and Genomics Core, National Institutes of Health, Bethesda, MD, USA
| | - Kimberly A Woodhouse
- DNA Sequencing and Genomics Core, National Institutes of Health, Bethesda, MD, USA
| | - Kahraman Tanriverdi
- Department of Medicine, University of Massachusetts Medical School, Worchester, MA, USA
| | - Jane E Freedman
- Department of Medicine, University of Massachusetts Medical School, Worchester, MA, USA
| | - Nalini Raghavachari
- National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Josée Dupuis
- The Framingham Heart Study and the Division of Intramural Research, National Heart, Lung and Blood Institute, National Institutes of Health, 73 Mt. Wayte Avenue, Suite 2, Framingham, MA, 01702, USA.,Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
| | - Andrew D Johnson
- The Framingham Heart Study and the Division of Intramural Research, National Heart, Lung and Blood Institute, National Institutes of Health, 73 Mt. Wayte Avenue, Suite 2, Framingham, MA, 01702, USA
| | - Christopher J O'Donnell
- The Framingham Heart Study and the Division of Intramural Research, National Heart, Lung and Blood Institute, National Institutes of Health, 73 Mt. Wayte Avenue, Suite 2, Framingham, MA, 01702, USA.,Cardiology Section, Department of Medicine, Boston VA Healthcare, Boston, MA, USA
| | - Daniel Levy
- The Framingham Heart Study and the Division of Intramural Research, National Heart, Lung and Blood Institute, National Institutes of Health, 73 Mt. Wayte Avenue, Suite 2, Framingham, MA, 01702, USA.
| | - Peter J Munson
- Mathematical and Statistical Computing Laboratory, Center for Information Technology, National Institutes of Health, Bethesda, MD, USA. .,National Institutes of Health, Bldg 12A, Room 2003, Bethesda, MD, 20892-5626, USA.
| |
Collapse
|
132
|
Guo L, Zhang Q, Ma X, Wang J, Liang T. miRNA and mRNA expression analysis reveals potential sex-biased miRNA expression. Sci Rep 2017; 7:39812. [PMID: 28045090 PMCID: PMC5206641 DOI: 10.1038/srep39812] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 11/25/2016] [Indexed: 12/22/2022] Open
Abstract
Recent studies suggest that mRNAs may be differentially expressed between males and females. This study aimed to perform expression analysis of mRNA and its main regulatory molecule, microRNA (miRNA), to discuss the potential sex-specific expression patterns using abnormal expression profiles from The Cancer Genome Atlas database. Generally, deregulated miRNAs and mRNAs had consistent expression between males and females, but some miRNAs may be oppositely expressed in specific diseases: up-regulated in one group and down-regulated in another. Studies of miRNA gene families and clusters further confirmed that these sequence or location related miRNAs might have opposing expression between sexes. The specific miRNA might have greater expression divergence across different groups, suggesting flexible expression across different individuals, especially in tumor samples. The typical analysis regardless of the sex will ignore or balance these sex-specific deregulated miRNAs. Compared with flexible miRNAs, their targets of mRNAs showed relative stable expression between males and females. These relevant results provide new insights into miRNA-mRNA interaction and sex difference.
Collapse
Affiliation(s)
- Li Guo
- Department of Bioinformatics, School of Geographic and Biologic Information, Nanjing University of Posts and Telecommunications, Nanjing, 210023, China
| | - Qiang Zhang
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Xiao Ma
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Jun Wang
- Department of Bioinformatics, School of Geographic and Biologic Information, Nanjing University of Posts and Telecommunications, Nanjing, 210023, China
| | - Tingming Liang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210023, China
| |
Collapse
|
133
|
Fejes Z, Póliska S, Czimmerer Z, Káplár M, Penyige A, Gál Szabó G, Beke Debreceni I, Kunapuli SP, Kappelmayer J, Nagy B. Hyperglycaemia suppresses microRNA expression in platelets to increase P2RY12 and SELP levels in type 2 diabetes mellitus. Thromb Haemost 2016; 117:529-542. [PMID: 27975100 DOI: 10.1160/th16-04-0322] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Accepted: 11/26/2016] [Indexed: 12/19/2022]
Abstract
Megakaryocyte (MK)-derived miRNAs have been detected in platelets. Here, we analysed the expression of platelet and circulating miR-223, miR-26b, miR-126 and miR-140 that might be altered with their target mRNAs in type 2 diabetes mellitus (DM2). MiRNAs were isolated from leukocyte-depleted platelets and plasma samples obtained from 28 obese DM2, 19 non-DM obese and 23 healthy individuals. The effect of hyperglycaemia on miRNAs was also evaluated in MKs using MEG-01 and K562 cells under hyperglycaemic conditions after 8 hours up to four weeks. Quantitation of mature miRNA, pre-miRNAs and target mRNA levels (P2RY12 and SELP) were measured by RT-qPCR. To prove the association of miR-26b and miR-140 with SELP (P-selectin) mRNA level, overexpression or inhibition of these miRNAs in MEG-01 MKs was performed using mimics or anti-miRNAs, respectively. The contribution of calpain substrate Dicer to modulation of miRNAs was studied by calpain inhibition. Platelet activation was evaluated via surface P-selectin by flow cytometry. Mature and pre-forms of investigated miRNAs were significantly reduced in DM2, and platelet P2RY12 and SELP mRNA levels were elevated by two-fold at increased platelet activation compared to controls. Significantly blunted miRNA expressions were observed by hyperglycaemia in MEG-01 and K562-MK cells versus baseline values, while the manipulation of miR-26b and miR-140 expression affected SELP mRNA level. Calpeptin pretreatment restored miRNA levels in hyperglycaemic MKs. Overall, miR-223, miR-26b, miR-126 and miR-140 are expressed at a lower level in platelets and MKs in DM2 causing upregulation of P2RY12 and SELP mRNAs that may contribute to adverse platelet function.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Béla Nagy
- Béla Nagy Jr, MD, PhD, Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98. H-4032, Debrecen, Hungary, Tel.: +36 52 340 006, Fax: +36 52 417 631, E-mail:
| |
Collapse
|
134
|
Abstract
Publisher's Note: This article has a companion Point by Brass et al. Publisher's Note: Join in the discussion of these articles at Blood Advances Community Conversations.
Collapse
|
135
|
Rocca B, Husted S. Safety of Antithrombotic Agents in Elderly Patients with Acute Coronary Syndromes. Drugs Aging 2016; 33:233-48. [PMID: 26941087 DOI: 10.1007/s40266-016-0359-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
There are unique challenges in the treatment and prevention of acute coronary syndromes (ACS) with antithrombotics in elderly patients: elderly patients usually require multiple drugs due to comorbidities, are highly susceptible to adverse drug reactions and drug-drug interactions, may have cognitive problems affecting compliance and complications, are especially exposed to the risk of falls and, most importantly, ageing is an independent risk factor for bleeding. Antithrombotic drugs, alone or in association, further and variously amplify age-related bleeding risk. Moreover, age-related changes in primary haemostasis may potentially affect the pharmacodynamics of some antiplatelet drugs. Thus, elderly subjects might be more or less sensitive to standard antiplatelet regimens depending on individual characteristics affecting antiplatelet drug response. Importantly, elderly patients are a rapidly growing population worldwide, have the highest incidence of ACS, but are poorly represented in clinical trials. As a consequence, evidence on antithrombotic drug benefits and risks is limited. Thus, in the real-world setting, older people are often denied antithrombotic drugs because of unjustified concerns, or might be over-treated and exposed to excessive bleeding risk. Personalized antithrombotic therapy in elderly patients is particularly critical, to minimize risks without affecting efficacy.
Collapse
Affiliation(s)
- Bianca Rocca
- Institute of Pharmacology, Catholic University School of Medicine, Largo F.Vito 1, 00168, Rome, Italy.
| | - Steen Husted
- Medical Department, Hospital Unit West, Herning/Holstebro and Institute of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
136
|
Kim J, Choi GH, Ko KH, Kim JO, Oh SH, Park YS, Kim OJ, Kim NK. Association of the Single Nucleotide Polymorphisms in microRNAs 130b, 200b, and 495 with Ischemic Stroke Susceptibility and Post-Stroke Mortality. PLoS One 2016; 11:e0162519. [PMID: 27603512 PMCID: PMC5014326 DOI: 10.1371/journal.pone.0162519] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 08/24/2016] [Indexed: 01/26/2023] Open
Abstract
The microRNA (miRNA) is a small non-coding RNA molecule that modulates gene expression at the posttranscriptional level. Platelets have a crucial role in both hemostasis and thrombosis, a condition that can occlude a cerebral artery and cause ischemic stroke. miR-130b, miR-200b, and miR-495 are potential genetic modulators involving platelet production and activation. We hypothesized that single nucleotide polymorphisms (SNPs) in these miRNAs might potentially contribute to the susceptibility to ischemic stroke and post-stroke mortality. This study included 523 ischemic stroke patients and 400 control subjects. We investigated the association of three miRNA SNPs (miR-130bT>C, miR-200bT>C, and miR-495A>C) with ischemic stroke prevalence and post-stroke mortality. In the multivariate logistic regression, there was no statistically significant difference in the distribution of miR-130bT>C, miR-200bT>C, or miR-495A>C between the ischemic stroke and control groups. In the subgroup analysis based on ischemic stroke subtype, the miR-200b CC genotype was less frequently found in the large-artery atherosclerosis stroke subtype compared with controls (TT+CT vs CC; adjusted odds ratio for CC, 0.506; 95% confidence interval, 0.265-0.965). During a mean follow-up period of 4.80 ± 2.11 years after stroke onset, there were 106 all-cause deaths among the 523 stroke patients. Multivariate Cox regression analysis did not find a significant association between post-stroke mortality and three miRNA SNPs. Our findings suggest that the functional SNP of miR-200b might be responsible for the susceptibility to large-artery atherosclerotic stroke.
Collapse
Affiliation(s)
- Jinkwon Kim
- Department of Neurology, CHA Bundang Medical Center, School of Medicine, CHA University, Seongnam-si, 13496, South Korea
| | - Gun Ho Choi
- Institute for Clinical Research, CHA Bundang Medical Center, School of Medicine, CHA University, Seongnam-si, 13496, South Korea
| | - Ki Han Ko
- Institute for Clinical Research, CHA Bundang Medical Center, School of Medicine, CHA University, Seongnam-si, 13496, South Korea
| | - Jung Oh Kim
- Institute for Clinical Research, CHA Bundang Medical Center, School of Medicine, CHA University, Seongnam-si, 13496, South Korea
| | - Seung Hun Oh
- Department of Neurology, CHA Bundang Medical Center, School of Medicine, CHA University, Seongnam-si, 13496, South Korea
| | - Young Seok Park
- Department of Neurosurgery, Chungbuk National University Hospital, Chungbuk National University, Cheongju-si, 28644, South Korea
| | - Ok Joon Kim
- Department of Neurology, CHA Bundang Medical Center, School of Medicine, CHA University, Seongnam-si, 13496, South Korea
- * E-mail: (NKK); (OJK)
| | - Nam Keun Kim
- Institute for Clinical Research, CHA Bundang Medical Center, School of Medicine, CHA University, Seongnam-si, 13496, South Korea
- Department of Biomedical Science, College of Life Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, 13488, South Korea
- * E-mail: (NKK); (OJK)
| |
Collapse
|
137
|
Yeung J, Tourdot BE, Adili R, Green AR, Freedman CJ, Fernandez-Perez P, Yu J, Holman TR, Holinstat M. 12(S)-HETrE, a 12-Lipoxygenase Oxylipin of Dihomo-γ-Linolenic Acid, Inhibits Thrombosis via Gαs Signaling in Platelets. Arterioscler Thromb Vasc Biol 2016; 36:2068-77. [PMID: 27470510 DOI: 10.1161/atvbaha.116.308050] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 07/15/2016] [Indexed: 01/09/2023]
Abstract
OBJECTIVE Dietary supplementation with polyunsaturated fatty acids has been widely used for primary and secondary prevention of cardiovascular disease in individuals at risk; however, the cardioprotective benefits of polyunsaturated fatty acids remain controversial because of lack of mechanistic and in vivo evidence. We present direct evidence that an omega-6 polyunsaturated fatty acid, dihomo-γ-linolenic acid (DGLA), exhibits in vivo cardioprotection through 12-lipoxygenase (12-LOX) oxidation of DGLA to its reduced oxidized lipid form, 12(S)-hydroxy-8Z,10E,14Z-eicosatrienoic acid (12(S)-HETrE), inhibiting platelet activation and thrombosis. APPROACH AND RESULTS DGLA inhibited ex vivo platelet aggregation and Rap1 activation in wild-type mice, but not in mice lacking 12-LOX expression (12-LOX(-/-)). Similarly, wild-type mice treated with DGLA were able to reduce thrombus growth (platelet and fibrin accumulation) after laser-induced injury of the arteriole of the cremaster muscle, but not 12-LOX(-/-) mice, supporting a 12-LOX requirement for mediating the inhibitory effects of DGLA on platelet-mediated thrombus formation. Platelet activation and thrombus formation were also suppressed when directly treated with 12(S)-HETrE. Importantly, 2 hemostatic models, tail bleeding and arteriole rupture of the cremaster muscle, showed no alteration in hemostasis after 12(S)-HETrE treatment. Finally, the mechanism for 12(S)-HETrE protection was shown to be mediated via a Gαs-linked G-protein-coupled receptor pathway in human platelets. CONCLUSIONS This study provides the direct evidence that an omega-6 polyunsaturated fatty acid, DGLA, inhibits injury-induced thrombosis through its 12-LOX oxylipin, 12(S)-HETrE, which strongly supports the potential cardioprotective benefits of DGLA supplementation through its regulation of platelet function. Furthermore, this is the first evidence of a 12-LOX oxylipin regulating platelet function in a Gs α subunit-linked G-protein-coupled receptor-dependent manner.
Collapse
Affiliation(s)
- Jennifer Yeung
- From the Department of Pharmacology (J.Y., B.E.T., R.A., M.H.) and Department of Internal Medicine, Division of Cardiovascular Medicine (M.H.), University of Michigan, Ann Arbor; Cardeza Foundation for Hematological Research, Thomas Jefferson University, Philadelphia, PA (J.Y., B.E.T., R.A., P.F.-P., J.Y., M.H.); and Department of Chemistry and Biochemistry, University of California Santa Cruz (A.R.G., C.J.F., T.R.H.)
| | - Benjamin E Tourdot
- From the Department of Pharmacology (J.Y., B.E.T., R.A., M.H.) and Department of Internal Medicine, Division of Cardiovascular Medicine (M.H.), University of Michigan, Ann Arbor; Cardeza Foundation for Hematological Research, Thomas Jefferson University, Philadelphia, PA (J.Y., B.E.T., R.A., P.F.-P., J.Y., M.H.); and Department of Chemistry and Biochemistry, University of California Santa Cruz (A.R.G., C.J.F., T.R.H.)
| | - Reheman Adili
- From the Department of Pharmacology (J.Y., B.E.T., R.A., M.H.) and Department of Internal Medicine, Division of Cardiovascular Medicine (M.H.), University of Michigan, Ann Arbor; Cardeza Foundation for Hematological Research, Thomas Jefferson University, Philadelphia, PA (J.Y., B.E.T., R.A., P.F.-P., J.Y., M.H.); and Department of Chemistry and Biochemistry, University of California Santa Cruz (A.R.G., C.J.F., T.R.H.)
| | - Abigail R Green
- From the Department of Pharmacology (J.Y., B.E.T., R.A., M.H.) and Department of Internal Medicine, Division of Cardiovascular Medicine (M.H.), University of Michigan, Ann Arbor; Cardeza Foundation for Hematological Research, Thomas Jefferson University, Philadelphia, PA (J.Y., B.E.T., R.A., P.F.-P., J.Y., M.H.); and Department of Chemistry and Biochemistry, University of California Santa Cruz (A.R.G., C.J.F., T.R.H.)
| | - Cody J Freedman
- From the Department of Pharmacology (J.Y., B.E.T., R.A., M.H.) and Department of Internal Medicine, Division of Cardiovascular Medicine (M.H.), University of Michigan, Ann Arbor; Cardeza Foundation for Hematological Research, Thomas Jefferson University, Philadelphia, PA (J.Y., B.E.T., R.A., P.F.-P., J.Y., M.H.); and Department of Chemistry and Biochemistry, University of California Santa Cruz (A.R.G., C.J.F., T.R.H.)
| | - Pilar Fernandez-Perez
- From the Department of Pharmacology (J.Y., B.E.T., R.A., M.H.) and Department of Internal Medicine, Division of Cardiovascular Medicine (M.H.), University of Michigan, Ann Arbor; Cardeza Foundation for Hematological Research, Thomas Jefferson University, Philadelphia, PA (J.Y., B.E.T., R.A., P.F.-P., J.Y., M.H.); and Department of Chemistry and Biochemistry, University of California Santa Cruz (A.R.G., C.J.F., T.R.H.)
| | - Johnny Yu
- From the Department of Pharmacology (J.Y., B.E.T., R.A., M.H.) and Department of Internal Medicine, Division of Cardiovascular Medicine (M.H.), University of Michigan, Ann Arbor; Cardeza Foundation for Hematological Research, Thomas Jefferson University, Philadelphia, PA (J.Y., B.E.T., R.A., P.F.-P., J.Y., M.H.); and Department of Chemistry and Biochemistry, University of California Santa Cruz (A.R.G., C.J.F., T.R.H.)
| | - Theodore R Holman
- From the Department of Pharmacology (J.Y., B.E.T., R.A., M.H.) and Department of Internal Medicine, Division of Cardiovascular Medicine (M.H.), University of Michigan, Ann Arbor; Cardeza Foundation for Hematological Research, Thomas Jefferson University, Philadelphia, PA (J.Y., B.E.T., R.A., P.F.-P., J.Y., M.H.); and Department of Chemistry and Biochemistry, University of California Santa Cruz (A.R.G., C.J.F., T.R.H.)
| | - Michael Holinstat
- From the Department of Pharmacology (J.Y., B.E.T., R.A., M.H.) and Department of Internal Medicine, Division of Cardiovascular Medicine (M.H.), University of Michigan, Ann Arbor; Cardeza Foundation for Hematological Research, Thomas Jefferson University, Philadelphia, PA (J.Y., B.E.T., R.A., P.F.-P., J.Y., M.H.); and Department of Chemistry and Biochemistry, University of California Santa Cruz (A.R.G., C.J.F., T.R.H.).
| |
Collapse
|
138
|
Zou S, Teixeira AM, Yin M, Xiang Y, Xavier-Ferrucio J, Zhang PX, Hwa J, Min W, Krause DS. Leukaemia-associated Rho guanine nucleotide exchange factor (LARG) plays an agonist specific role in platelet function through RhoA activation. Thromb Haemost 2016; 116:506-16. [PMID: 27345948 DOI: 10.1160/th15-11-0848] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 05/14/2016] [Indexed: 11/05/2022]
Abstract
Leukemia-Associated RhoGEF (LARG) is highly expressed in platelets, which are essential for maintaining normal haemostasis. We studied the function of LARG in murine and human megakaryocytes and platelets with Larg knockout (KO), shRNA-mediated knockdown and small molecule-mediated inhibition. We found that LARG is important for human, but not murine, megakaryocyte maturation. Larg KO mice exhibit macrothrombocytopenia, internal bleeding in the ovaries and prolonged bleeding times. KO platelets have impaired aggregation, α-granule release and integrin α2bβ3 activation in response to thrombin and thromboxane, but not to ADP. The same agonist-specific reductions in platelet aggregation occur in human platelets treated with a LARG inhibitor. Larg KO platelets have reduced RhoA activation and myosin light chain phosphorylation, suggesting that Larg plays an agonist-specific role in platelet signal transduction. Using two different in vivo assays, Larg KO mice are protected from in vivo thrombus formation. Together, these results establish that LARG regulates human megakaryocyte maturation, and is critical for platelet function in both humans and mice.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Diane S Krause
- Diane S. Krause, Yale Stem Cell Center, 10 Amistad Street, Room 214I, New Haven, CT 06509, USA, Tel.: +1 203 785 7089, Fax: +1 203 785 4305, E-mail:
| |
Collapse
|
139
|
Khatlani T, Pradhan S, Da Q, Shaw T, Buchman VL, Cruz MA, Vijayan KV. A Novel Interaction of the Catalytic Subunit of Protein Phosphatase 2A with the Adaptor Protein CIN85 Suppresses Phosphatase Activity and Facilitates Platelet Outside-in αIIbβ3 Integrin Signaling. J Biol Chem 2016; 291:17360-8. [PMID: 27334924 DOI: 10.1074/jbc.m115.704296] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Indexed: 11/06/2022] Open
Abstract
The transduction of signals generated by protein kinases and phosphatases are critical for the ability of integrin αIIbβ3 to support stable platelet adhesion and thrombus formation. Unlike kinases, it remains unclear how serine/threonine phosphatases engage the signaling networks that are initiated following integrin ligation. Because protein-protein interactions form the backbone of signal transduction, we searched for proteins that interact with the catalytic subunit of protein phosphatase 2A (PP2Ac). In a yeast two-hybrid study, we identified a novel interaction between PP2Ac and an adaptor protein CIN85 (Cbl-interacting protein of 85 kDa). Truncation and alanine mutagenesis studies revealed that PP2Ac binds to the P3 block ((396)PAIPPKKPRP(405)) of the proline-rich region in CIN85. The interaction of purified PP2Ac with CIN85 suppressed phosphatase activity. Human embryonal kidney 293 αIIbβ3 cells overexpressing a CIN85 P3 mutant, which cannot support PP2Ac binding, displayed decreased adhesion to immobilized fibrinogen. Platelets contain the ∼85 kDa CIN85 protein along with the PP2Ac-CIN85 complex. A myristylated cell-permeable peptide derived from residues 395-407 of CIN85 protein (P3 peptide) disrupted the platelet PP2Ac-CIN85 complex and decreased αIIbβ3 signaling dependent functions such as platelet spreading on fibrinogen and thrombin-mediated fibrin clot retraction. In a phospho-profiling study P3 peptide treated platelets also displayed decreased phosphorylation of several signaling proteins including Src and GSK3β. Taken together, these data support a role for the novel PP2Ac-CIN85 complex in supporting integrin-dependent platelet function by dampening the phosphatase activity.
Collapse
Affiliation(s)
| | | | - Qi Da
- From the Departments of Medicine
| | | | - Vladimir L Buchman
- the School of Biosciences, Cardiff University, Wales CF10 3AX, United Kingdom
| | - Miguel A Cruz
- From the Departments of Medicine, Pediatrics, and Molecular Physiology and Biophysics, Baylor College of Medicine and Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey Veterans Affairs Medical Center (MEDVAMC), Houston, Texas 77030 and
| | - K Vinod Vijayan
- From the Departments of Medicine, Pediatrics, and Molecular Physiology and Biophysics, Baylor College of Medicine and Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey Veterans Affairs Medical Center (MEDVAMC), Houston, Texas 77030 and
| |
Collapse
|
140
|
Nagalla S, Bray PF. Personalized medicine in thrombosis: back to the future. Blood 2016; 127:2665-71. [PMID: 26847245 PMCID: PMC4891951 DOI: 10.1182/blood-2015-11-634832] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Accepted: 01/31/2016] [Indexed: 01/26/2023] Open
Abstract
Most physicians believe they practiced personalized medicine prior to the genomics era that followed the sequencing of the human genome. The focus of personalized medicine has been primarily genomic medicine, wherein it is hoped that the nucleotide dissimilarities among different individuals would provide clinicians with more precise understanding of physiology, more refined diagnoses, better disease risk assessment, earlier detection and monitoring, and tailored treatments to the individual patient. However, to date, the "genomic bench" has not worked itself to the clinical thrombosis bedside. In fact, traditional plasma-based hemostasis-thrombosis laboratory testing, by assessing functional pathways of coagulation, may better help manage venous thrombotic disease than a single DNA variant with a small effect size. There are some new and exciting discoveries in the genetics of platelet reactivity pertaining to atherothrombotic disease. Despite a plethora of genetic/genomic data on platelet reactivity, there are relatively little actionable pharmacogenetic data with antiplatelet agents. Nevertheless, it is crucial for genome-wide DNA/RNA sequencing to continue in research settings for causal gene discovery, pharmacogenetic purposes, and gene-gene and gene-environment interactions. The potential of genomics to advance medicine will require integration of personal data that are obtained in the patient history: environmental exposures, diet, social data, etc. Furthermore, without the ritual of obtaining this information, we will have depersonalized medicine, which lacks the precision needed for the research required to eventually incorporate genomics into routine, optimal, and value-added clinical care.
Collapse
Affiliation(s)
- Srikanth Nagalla
- The Cardeza Foundation for Hematologic Research and the Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA
| | - Paul F Bray
- The Cardeza Foundation for Hematologic Research and the Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA
| |
Collapse
|
141
|
Vélez P, Ocaranza-Sánchez R, López-Otero D, Grigorian-Shamagian L, Rosa I, Bravo SB, González-Juanatey JR, García Á. 2D-DIGE-based proteomic analysis of intracoronary versus peripheral arterial blood platelets from acute myocardial infarction patients: Upregulation of platelet activation biomarkers at the culprit site. Proteomics Clin Appl 2016; 10:851-8. [DOI: 10.1002/prca.201500120] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 04/01/2016] [Accepted: 04/14/2016] [Indexed: 11/09/2022]
Affiliation(s)
- Paula Vélez
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS); Universidade de Santiago de Compostela; Santiago de Compostela Spain
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS); Santiago de Compostela Spain
| | - Raymundo Ocaranza-Sánchez
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS); Santiago de Compostela Spain
- Cardiology Department and Coronary Care Unit; Hospital Clínico Universitario de Santiago; Santiago de Compostela Spain
| | - Diego López-Otero
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS); Santiago de Compostela Spain
- Cardiology Department and Coronary Care Unit; Hospital Clínico Universitario de Santiago; Santiago de Compostela Spain
| | | | - Isaac Rosa
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS); Universidade de Santiago de Compostela; Santiago de Compostela Spain
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS); Santiago de Compostela Spain
| | - Susana Belén Bravo
- Proteomic Unit, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS); Hospital Clínico Universitario de Santiago; Santiago de Compostela Spain
| | - José Ramón González-Juanatey
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS); Santiago de Compostela Spain
- Cardiology Department and Coronary Care Unit; Hospital Clínico Universitario de Santiago; Santiago de Compostela Spain
| | - Ángel García
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS); Universidade de Santiago de Compostela; Santiago de Compostela Spain
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS); Santiago de Compostela Spain
| |
Collapse
|
142
|
Guo L, Liang T, Yu J, Zou Q. A Comprehensive Analysis of miRNA/isomiR Expression with Gender Difference. PLoS One 2016; 11:e0154955. [PMID: 27167065 PMCID: PMC4864079 DOI: 10.1371/journal.pone.0154955] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 04/21/2016] [Indexed: 01/09/2023] Open
Abstract
Although microRNAs (miRNAs) have been widely studied as epigenetic regulation molecules, fewer studies focus on the gender difference at the miRNA and isomiR expression levels. In this study, we aim to understand the potential relationships between gender difference and miRNA/isomiR expression through a comprehensive analysis of small RNA-sequencing datasets based on different human diseases and tissues. Based on specific samples from males and females, we determined that some miRNAs may be diversely expressed between different tissues and genders. Thus, these miRNAs may exhibit inconsistent and even opposite expression between males and females. According to deregulated miRNA expression profiles, some dominantly expressed miRNA loci were selected to analyze isomiR expression patterns using rates of dominant isomiRs. In some miRNA loci, isomiRs showed statistical significance between tumor and normal samples and between males and females samples, suggesting that isomiR expression patterns are not always invariable but may vary between males and females, as well as among different tissues, tumors, and normal samples. The divergence implicates the fluctuation in the expression of miRNA and its detailed expression at the isomiR levels. The divergence also indicates that gender difference may be an important factor that affects the screening of disease-associated miRNAs and isomiRs. This study suggests that miRNA/isomiR expression and gender difference may be more complex than previously assumed and should be further studied according to specific samples from males or females.
Collapse
Affiliation(s)
- Li Guo
- Department of Bioinformatics, School of Geographic and Biologic Information, Nanjing University of Posts and Telecommunications, Nanjing, China
- * E-mail: (LG); (QZ)
| | - Tingming Liang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Jiafeng Yu
- Shandong Provincial Key Laboratory of Functional Macromolecular Biophysics, Institute of Biophysics, Dezhou University, Dezhou, China
| | - Quan Zou
- School of Computer Science and Technology, Tianjin University, Tianjin, China
- State Key Laboratory of Medicinal Chemical Biology, NanKai University, Tianjin, China
- * E-mail: (LG); (QZ)
| |
Collapse
|
143
|
Simon L, Chen E, Edelstein L, Kong X, Bhatlekar S, Rigoutsos I, Bray P, Shaw C. Integrative Multi-omic Analysis of Human Platelet eQTLs Reveals Alternative Start Site in Mitofusin 2. Am J Hum Genet 2016; 98:883-897. [PMID: 27132591 DOI: 10.1016/j.ajhg.2016.03.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 03/11/2016] [Indexed: 02/07/2023] Open
Abstract
Platelets play a central role in ischemic cardiovascular events. Cardiovascular disease (CVD) is a major cause of death worldwide. Numerous genome-wide association studies (GWASs) have identified loci associated with CVD risk. However, our understanding of how these variants contribute to disease is limited. Using data from the platelet RNA and expression 1 (PRAX1) study, we analyzed cis expression quantitative trait loci (eQTLs) in platelets from 154 normal human subjects. We confirmed these results in silico by performing allele-specific expression (ASE) analysis, which demonstrated that the allelic directionality of eQTLs and ASE patterns correlate significantly. Comparison of platelet eQTLs with data from the Genotype-Tissue Expression (GTEx) project revealed that a number of platelet eQTLs are platelet specific and that platelet eQTL peaks localize to the gene body at a higher rate than eQTLs from other tissues. Upon integration with data from previously published GWASs, we found that the trait-associated variant rs1474868 coincides with the eQTL peak for mitofusin 2 (MFN2). Additional experimental and computational analyses revealed that this eQTL is linked to an unannotated alternate MFN2 start site preferentially expressed in platelets. Integration of phenotype data from the PRAX1 study showed that MFN2 expression levels were significantly associated with platelet count. This study links the variant rs1474868 to a platelet-specific regulatory role for MFN2 and demonstrates the utility of integrating multi-omic data with eQTL analysis in disease-relevant tissues for interpreting GWAS results.
Collapse
|
144
|
Abstract
There are clear age-related changes in platelet count and function, driven by changes in hematopoietic tissue, the composition of the blood and vascular health. Platelet count remains relatively stable during middle age (25–60 years old) but falls in older people. The effect of age on platelet function is slightly less clear. The longstanding view is that platelet reactivity increases with age in an almost linear fashion. There are, however, serious limitations to the data supporting this dogma. We can conclude that platelet function increases during middle age, but little evidence exists on the changes in platelet responsiveness in old age (>75 years old). This change in platelet function is driven by differential mRNA and microRNA expression, an increase in oxidative stress and changes in platelet receptors. These age-related changes in platelets are particularly pertinent given that thrombotic disease and use of anti-platelet drugs is much more prevalent in the elderly population, yet the majority of platelet research is carried out in young to middle-aged (20–50 years old) human volunteers and young mice (2–6 months old). We know relatively little about exactly how platelets from people over 75 years old differ from those of middle-aged subjects, and we know even less about the mechanisms that drive these changes. Addressing these gaps in our knowledge will provide substantial understanding in how cell signalling changes during ageing and will enable the development of more precise anti-platelet therapies.
Collapse
Affiliation(s)
- Chris I Jones
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Harborne Building, Whiteknights, Reading, Berkshire, RG6 6AS, UK.
| |
Collapse
|
145
|
Abstract
Platelets are megakaryocyte-derived cellular fragments, which lack a nucleus and are the smallest circulating cells and are classically known to have a major role in supporting hemostasis. Apart from this well-established role, it is now becoming evident that platelets are also capable of conveying other important functions, such as during infection and inflammation. This paper will outline these nonhemostatic functions in two major sections termed "Platelets versus pathogens" and "Platelet-target cell communication". Platelets actively contribute to protection against invading pathogens and are capable of regulating immune functions in various target cells, all through sophisticated and efficient mechanisms. These relatively novel features will be highlighted, illustrating the multifunctional role of platelets in inflammation.
Collapse
Affiliation(s)
- Rick Kapur
- Toronto Platelet Immunobiology Group, Keenan Research Centre for Biomedical Science, St. Michael׳s Hospital, Canadian Blood Services, Toronto, Ontario, Canada
| | - John W Semple
- Toronto Platelet Immunobiology Group, Keenan Research Centre for Biomedical Science, St. Michael׳s Hospital, Canadian Blood Services, Toronto, Ontario, Canada; Departments of Pharmacology, Medicine, and Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
146
|
New molecular insights into modulation of platelet reactivity in aspirin-treated patients using a network-based approach. Hum Genet 2016; 135:403-414. [DOI: 10.1007/s00439-016-1642-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 01/23/2016] [Indexed: 10/22/2022]
|
147
|
Best MG, Sol N, Tannous BA, Wesseling P, Wurdinger T. Re: a Word of Caution on New and Revolutionary Diagnostic Tests. Cancer Cell 2016; 29:143-4. [PMID: 26859454 DOI: 10.1016/j.ccell.2016.01.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 01/11/2016] [Accepted: 01/11/2016] [Indexed: 11/24/2022]
Affiliation(s)
- Myron G Best
- Department of Pathology, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands; Department of Neurosurgery, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
| | - Nik Sol
- Department of Neurology, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
| | - Bakhos A Tannous
- Department of Neurology, Massachusetts General Hospital and Neuroscience Program, Harvard Medical School, 149 13th Street, Charlestown, MA 02129, USA
| | - Pieter Wesseling
- Department of Pathology, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands; Department of Pathology, Radboud University Medical Center, 6500 HB Nijmegen, the Netherlands
| | - Thomas Wurdinger
- Department of Neurosurgery, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands; Department of Neurology, Massachusetts General Hospital and Neuroscience Program, Harvard Medical School, 149 13th Street, Charlestown, MA 02129, USA; thromboDx B.V., 1098 EA Amsterdam, the Netherlands.
| |
Collapse
|
148
|
Potential Diagnostic and Prognostic Biomarkers of Epigenetic Drift within the Cardiovascular Compartment. BIOMED RESEARCH INTERNATIONAL 2016; 2016:2465763. [PMID: 26942189 PMCID: PMC4749768 DOI: 10.1155/2016/2465763] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 11/02/2015] [Accepted: 11/24/2015] [Indexed: 12/15/2022]
Abstract
Biomarkers encompass a wide range of different measurable indicators, representing a tangible link to physiological changes occurring within the body. Accessibility, sensitivity, and specificity are significant factors in biomarker suitability. New biomarkers continue to be discovered, and questions over appropriate selection and assessment of their usefulness remain. If traditional markers of inflammation are not sufficiently robust in their specificity, then perhaps alternative means of detection may provide more information. Epigenetic drift (epigenetic modifications as they occur as a direct function with age), and its ancillary elements, including platelets, secreted microvesicles (MVs), and microRNA (miRNA), may hold enormous predictive potential. The majority of epigenetic drift observed in blood is independent of variations in blood cell composition, addressing concerns affecting traditional blood-based biomarker efficacy. MVs are found in plasma and other biological fluids in healthy individuals. Altered MV/miRNA profiles may also be found in individuals with various diseases. Platelets are also highly reflective of physiological and lifestyle changes, making them extremely sensitive biomarkers of human health. Platelets release increased levels of MVs in response to various stimuli and under a plethora of disease states, which demonstrate a functional effect on other cell types.
Collapse
|
149
|
Best MG, Sol N, Kooi I, Tannous J, Westerman BA, Rustenburg F, Schellen P, Verschueren H, Post E, Koster J, Ylstra B, Ameziane N, Dorsman J, Smit EF, Verheul HM, Noske DP, Reijneveld JC, Nilsson RJA, Tannous BA, Wesseling P, Wurdinger T. RNA-Seq of Tumor-Educated Platelets Enables Blood-Based Pan-Cancer, Multiclass, and Molecular Pathway Cancer Diagnostics. Cancer Cell 2015; 28:666-676. [PMID: 26525104 PMCID: PMC4644263 DOI: 10.1016/j.ccell.2015.09.018] [Citation(s) in RCA: 607] [Impact Index Per Article: 60.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Revised: 07/02/2015] [Accepted: 09/25/2015] [Indexed: 12/12/2022]
Abstract
Tumor-educated blood platelets (TEPs) are implicated as central players in the systemic and local responses to tumor growth, thereby altering their RNA profile. We determined the diagnostic potential of TEPs by mRNA sequencing of 283 platelet samples. We distinguished 228 patients with localized and metastasized tumors from 55 healthy individuals with 96% accuracy. Across six different tumor types, the location of the primary tumor was correctly identified with 71% accuracy. Also, MET or HER2-positive, and mutant KRAS, EGFR, or PIK3CA tumors were accurately distinguished using surrogate TEP mRNA profiles. Our results indicate that blood platelets provide a valuable platform for pan-cancer, multiclass cancer, and companion diagnostics, possibly enabling clinical advances in blood-based "liquid biopsies".
Collapse
Affiliation(s)
- Myron G Best
- Department of Pathology, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands; Department of Neurosurgery, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
| | - Nik Sol
- Department of Neurology, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
| | - Irsan Kooi
- Department of Clinical Genetics, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
| | - Jihane Tannous
- Department of Neurology, Massachusetts General Hospital and Neuroscience Program, Harvard Medical School, 149 13th Street, Charlestown, MA 02129, USA
| | - Bart A Westerman
- Department of Neurosurgery, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
| | - François Rustenburg
- Department of Pathology, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands; Department of Neurosurgery, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
| | - Pepijn Schellen
- Department of Neurosurgery, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands; thromboDx B.V., 1098 EA Amsterdam, the Netherlands
| | - Heleen Verschueren
- Department of Neurosurgery, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands; thromboDx B.V., 1098 EA Amsterdam, the Netherlands
| | - Edward Post
- Department of Neurosurgery, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands; thromboDx B.V., 1098 EA Amsterdam, the Netherlands
| | - Jan Koster
- Department of Oncogenomics, Academic Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Bauke Ylstra
- Department of Pathology, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
| | - Najim Ameziane
- Department of Clinical Genetics, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
| | - Josephine Dorsman
- Department of Clinical Genetics, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
| | - Egbert F Smit
- Department of Pulmonary Diseases, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
| | - Henk M Verheul
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
| | - David P Noske
- Department of Neurosurgery, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
| | - Jaap C Reijneveld
- Department of Neurology, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
| | - R Jonas A Nilsson
- Department of Neurosurgery, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands; thromboDx B.V., 1098 EA Amsterdam, the Netherlands; Department of Radiation Sciences, Oncology, Umeå University, 90185 Umeå, Sweden
| | - Bakhos A Tannous
- Department of Neurology, Massachusetts General Hospital and Neuroscience Program, Harvard Medical School, 149 13th Street, Charlestown, MA 02129, USA
| | - Pieter Wesseling
- Department of Pathology, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands; Department of Pathology, Radboud University Medical Center, 6500 HB Nijmegen, the Netherlands
| | - Thomas Wurdinger
- Department of Neurosurgery, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands; Department of Neurology, Massachusetts General Hospital and Neuroscience Program, Harvard Medical School, 149 13th Street, Charlestown, MA 02129, USA; thromboDx B.V., 1098 EA Amsterdam, the Netherlands.
| |
Collapse
|
150
|
Anti-miR-148a regulates platelet FcγRIIA signaling and decreases thrombosis in vivo in mice. Blood 2015; 126:2871-81. [PMID: 26516227 DOI: 10.1182/blood-2015-02-631135] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 10/21/2015] [Indexed: 12/27/2022] Open
Abstract
Fc receptor for IgG IIA (FcγRIIA)-mediated platelet activation is essential in heparin-induced thrombocytopenia (HIT) and other immune-mediated thrombocytopenia and thrombosis disorders. There is considerable interindividual variation in platelet FcγRIIA activation, the reasons for which remain unclear. We hypothesized that genetic variations between FcγRIIA hyper- and hyporesponders regulate FcγRIIA-mediated platelet reactivity and influence HIT susceptibility. Using unbiased genome-wide expression profiling, we observed that human hyporesponders to FcγRIIA activation showed higher platelet T-cell ubiquitin ligand-2 (TULA-2) mRNA expression than hyperresponders. Silent interfering RNA-mediated knockdown of TULA-2 resulted in hyperphosphorylation of spleen tyrosine kinase following FcγRIIA activation in HEL cells. Significantly, we found miR-148a-3p targeted and inhibited both human and mouse TULA-2 mRNA. Inhibition of miR-148a in FcγRIIA transgenic mice upregulated the TULA-2 level and reduced FcγRIIA- and glycoprotein VI-mediated platelet αIIbβ3 activation and calcium mobilization. Anti-miR-148a also reduced thrombus formation following intravascular platelet activation via FcγRIIA. These results show that TULA-2 is a target of miR-148a-3p, and TULA-2 serves as a negative regulator of FcγRIIA-mediated platelet activation. This is also the first study to show the effects of in vivo miRNA inhibition on platelet reactivity. Our work suggests that modulating miR-148a expression is a potential therapeutic approach for thrombosis.
Collapse
|