51
|
The Importance of Platelet Glycoside Residues in the Haemostasis of Patients with Immune Thrombocytopaenia. J Clin Med 2021; 10:jcm10081661. [PMID: 33924503 PMCID: PMC8069668 DOI: 10.3390/jcm10081661] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/26/2021] [Accepted: 04/09/2021] [Indexed: 01/14/2023] Open
Abstract
Loss of sialic acid from the carbohydrate side chains of platelet glycoproteins can affect platelet clearance, a proposed mechanism involved in the etiopathogenesis of immune thrombocytopaenia (ITP). We aimed to assess whether changes in platelet glycosylation in patients with ITP affected platelet counts, function, and apoptosis. This observational, prospective, and transversal study included 82 patients with chronic primary ITP and 115 healthy controls. We measured platelet activation markers and assayed platelet glycosylation and caspase activity, analysing samples using flow cytometry. Platelets from patients with ITP with a platelet count <30 × 103/µL presented less sialic acid. Levels of α1,6-fucose (a glycan residue that can directly regulate antibody-dependent cellular cytotoxicity) and α-mannose (which can be recognised by mannose-binding-lectin and activate the complement pathway) were increased in the platelets from these patients. Platelet surface exposure of other glycoside residues due to sialic acid loss inversely correlated with platelet count and the ability to be activated. Moreover, loss of sialic acid induced the ingestion of platelets by human hepatome HepG2 cells. Changes in glycoside composition of glycoproteins on the platelets’ surface impaired their functional capacity and increased their apoptosis. These changes in platelet glycoside residues appeared to be related to ITP severity.
Collapse
|
52
|
Ma X, Li Y, Kondo Y, Shi H, Han J, Jiang Y, Bai X, Archer-Hartmann SA, Azadi P, Ruan C, Fu J, Xia L. Slc35a1 deficiency causes thrombocytopenia due to impaired megakaryocytopoiesis and excessive platelet clearance in the liver. Haematologica 2021; 106:759-769. [PMID: 32303557 PMCID: PMC7927894 DOI: 10.3324/haematol.2019.225987] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Indexed: 12/27/2022] Open
Abstract
Sialic acid is a common terminal residue of glycans on proteins and
acidic sphingolipids such as gangliosides and has important biological
functions. The sialylation process is controlled by more than 20 different
sialyltransferases, many of which exhibit overlapping functions.
Thus, it is difficult to determine the overall biological function of sialylation
by targeted deletion of individual sialyltransferases. To address this
issue, we established a mouse line with the Slc35a1 gene flanked by loxP
sites. Slc35a1 encodes the cytidine-5’-monophosphate (CMP)-sialic acid
transporter that transports CMP-sialic acid from the cytoplasm into the
Golgi apparatus for sialylation. Here we report our study regarding the role
of sialylation on megakaryocytes and platelets using a mouse line with significantly
reduced sialylation in megakaryocytes and platelets (Plt Slc35a1–
/–). The major phenotype of Plt Slc35a1–/– mice was thrombocytopenia. The
number of bone marrow megakaryocytes in Plt Slc35a1–/– mice was
reduced, and megakaryocyte maturation was also impaired. In addition, an
increased number of desialylated platelets was cleared by Küpffer cells in
the liver of Plt Slc35a1–/– mice. This study provides new insights into the
role of sialylation in platelet homeostasis and the mechanisms of thrombocytopenia
in diseases associated with platelet desialylation, such as
immune thrombocytopenia and a rare congenital disorder of glycosylation
(CDG), SLC35A1-CDG, which is caused by SLC35A1 mutations.
Collapse
Affiliation(s)
- Xiaolin Ma
- Jiangsu Institute of Hematology, NHC Key Laboratory of Thrombosis and Hemostasis, The First Affiliated Hospital of Soochow University, Suzhou, China,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China,Department of Hematology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yun Li
- Jiangsu Institute of Hematology, NHC Key Laboratory of Thrombosis and Hemostasis, The First Affiliated Hospital of Soochow University, Suzhou, China,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Yuji Kondo
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Huiping Shi
- Jiangsu Institute of Hematology, NHC Key Laboratory of Thrombosis and Hemostasis, The First Affiliated Hospital of Soochow University, Suzhou, China,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China,Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Jingjing Han
- Jiangsu Institute of Hematology, NHC Key Laboratory of Thrombosis and Hemostasis, The First Affiliated Hospital of Soochow University, Suzhou, China,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Yizhi Jiang
- Jiangsu Institute of Hematology, NHC Key Laboratory of Thrombosis and Hemostasis, The First Affiliated Hospital of Soochow University, Suzhou, China,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Xia Bai
- Jiangsu Institute of Hematology, NHC Key Laboratory of Thrombosis and Hemostasis, The First Affiliated Hospital of Soochow University, Suzhou, China,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China,State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| | | | - Parastoo Azadi
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
| | - Changgeng Ruan
- Jiangsu Institute of Hematology, NHC Key Laboratory of Thrombosis and Hemostasis, The First Affiliated Hospital of Soochow University, Suzhou, China,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China,State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| | - Jianxin Fu
- Jiangsu Institute of Hematology, NHC Key Laboratory of Thrombosis and Hemostasis, The First Affiliated Hospital of Soochow University, Suzhou, China,Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA,Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lijun Xia
- Jiangsu Institute of Hematology, NHC Key Laboratory of Thrombosis and Hemostasis, The First Affiliated Hospital of Soochow University, Suzhou, China,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China,Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| |
Collapse
|
53
|
Xiang B, Zhang G, Zhang Y, Wu C, Joshi S, Morris AJ, Ware J, Smyth SS, Whiteheart SW, Li Z. Calcium Ion Chelation Preserves Platelet Function During Cold Storage. Arterioscler Thromb Vasc Biol 2021; 41:234-249. [PMID: 33176450 PMCID: PMC8158249 DOI: 10.1161/atvbaha.120.314879] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Platelet transfusion is a life-saving therapy to prevent or treat bleeding in patients with thrombocytopenia or platelet dysfunction. However, for >6 decades, safe and effective strategies for platelet storage have been an impediment to widespread use of platelet transfusion. Refrigerated platelets are cleared rapidly from circulation, precluding cold storage of platelets for transfusion. Consequently, platelets are stored at room temperature with an upper limit of 5 days due to risks of bacterial contamination and loss of platelet function. This practice severely limits platelet availability for transfusion. This study is to identify the mechanism of platelet clearance after cold storage and develop a method for platelet cold storage. Approach and Results: We found that rapid clearance of cold-stored platelets was largely due to integrin activation and apoptosis. Deficiency of integrin β3 or caspase-3 prolonged cold-stored platelets in circulation. Pretreatment of platelets with EGTA, a cell impermeable calcium ion chelator, reversely inhibited cold storage-induced platelet activation and consequently prolonged circulation of cold-stored platelets. Moreover, transfusion of EGTA-treated, cold-stored platelets, but not room temperature-stored platelets, into the mice deficient in glycoprotein Ibα significantly shortened tail-bleeding times and diminished blood loss. CONCLUSIONS Integrin activation and apoptosis is the underlying mechanism of rapid clearance of platelets after cold storage. Addition of a cell impermeable calcium ion chelator to platelet products is potentially a simple and effective method to enable cold storage of platelets for transfusion.
Collapse
Affiliation(s)
- Binggang Xiang
- Gill Heart and Vascular Institute, Division of Cardiovascular Medicine, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Guoying Zhang
- Gill Heart and Vascular Institute, Division of Cardiovascular Medicine, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Yan Zhang
- Gill Heart and Vascular Institute, Division of Cardiovascular Medicine, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Congqing Wu
- Gill Heart and Vascular Institute, Division of Cardiovascular Medicine, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Smita Joshi
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | - Andrew J. Morris
- Gill Heart and Vascular Institute, Division of Cardiovascular Medicine, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Jerry Ware
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Susan S. Smyth
- Gill Heart and Vascular Institute, Division of Cardiovascular Medicine, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
- Veterans Affairs Medical Center, Lexington, KY 40536, USA
| | - Sidney W. Whiteheart
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | - Zhenyu Li
- Gill Heart and Vascular Institute, Division of Cardiovascular Medicine, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
54
|
Wang Y, Chen W, Zhang W, Lee-Sundlov MM, Casari C, Berndt MC, Lanza F, Bergmeier W, Hoffmeister KM, Zhang XF, Li R. Desialylation of O-glycans on glycoprotein Ibα drives receptor signaling and platelet clearance. Haematologica 2021; 106:220-229. [PMID: 31974202 PMCID: PMC7776245 DOI: 10.3324/haematol.2019.240440] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Accepted: 01/22/2020] [Indexed: 11/16/2022] Open
Abstract
During infection neuraminidase desialylates platelets and induces their rapid clearance from circulation. The underlying molecular basis, particularly the role of platelet glycoprotein (GP)Ibα therein, is not clear. Utilizing genetically altered mice, we report that the extracellular domain of GPIbα, but neither von Willebrand factor nor ADAM17 (a disintegrin and metalloprotease 17), is required for platelet clearance induced by intravenous injection of neuraminidase. Lectin binding to platelet following neuraminidase injection over time revealed that the extent of desialylation of O-glycans correlates with the decrease of platelet count in mice. Injection of α2,3-neuraminidase reduces platelet counts in wild-type but not in transgenic mice expressing only a chimeric GPIbα that misses most of its extracellular domain. Neuraminidase treatment induces unfolding of the O-glycosylated mechanosensory domain in GPIbα as monitored by single-molecule force spectroscopy, increases the exposure of the ADAM17 shedding cleavage site in the mechanosensory domain on the platelet surface, and induces ligand-independent GPIb-IX signaling in human and murine platelets. These results suggest that desialylation of O-glycans of GPIbα induces unfolding of the mechanosensory domain, subsequent GPIb-IX signaling including amplified desialylation of N-glycans, and eventually rapid platelet clearance. This new molecular mechanism of GPIbα-facilitated clearance could potentially resolve many puzzling and seemingly contradicting observations associated with clearance of desialylated or hyposialylated platelet.
Collapse
Affiliation(s)
- Yingchun Wang
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| | - Wenchun Chen
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| | - Wei Zhang
- Department of Bioengineering, Lehigh University, Bethlehem, PA
| | | | - Caterina Casari
- McAllister Heart Institute, University of North Carolina, School of Medicine, Chapel Hill, NC
| | | | - Francois Lanza
- Université de Strasbourg, EFS-Alsace, Strasbourg, France
| | | | | | - X Frank Zhang
- Department of Bioengineering, Lehigh University, Bethlehem, PA
| | - Renhao Li
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| |
Collapse
|
55
|
N-acetylcysteine reduce the stress induced by cold storage of platelets: A potential way to extend shelf life of platelets. Transfus Apher Sci 2020; 60:103039. [PMID: 33388248 DOI: 10.1016/j.transci.2020.103039] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 11/26/2020] [Accepted: 12/01/2020] [Indexed: 10/22/2022]
Abstract
The room temperature storage used for platelets worldwide leads to platelet storage lesion (PSL) and risk of bacterial growth, limiting platelet shelf life and safety in transfusion. Thus, there is a need for an alternative storage method that can serve as effective temperature storage for platelet concentrates (PCs). In the previous investigation, we have shown that N-acetylcysteine (NAC) is a potential candidate for an additive solution to retain platelet characteristics during cold storage for up to 5 days. However, the study partially describes the efficacy and has drawbacks to address. Here, we used the apheresis platelet product with 50 mM NAC and stored up to 10 days under refrigerated condition (4 ± 1 °C). Stored platelet concentrates were analyzed for critical parameters such as platelet activation, annexin V binding, sialic acid, reactive oxygen species (ROS), neuraminidase activity, and in vivo efficacy using Prkdcscid mice. Investigation observations revealed that PCs with NAC showed reduced platelet activation, annexin V binding, ROS production, and sialic acid levels. in vivo recovery of PCs showed similar recovery rates stored PCs irrespective of treatment or storage condition. However, on the tenth day after 24 h, recovery in room temperature stored concentrates was about 32 %, whereas in NAC treated refrigerated concentrates, it stands at 47 %. These observations indicate that NAC addition protects refrigerated concentrates during long-term storage retaining the platelet integrity. The study also suggests that extending PC storage beyond 10 days is practically accomplishable with efficacy similar to room temperature (RT) stored PCs.
Collapse
|
56
|
Hegde S, Wellendorf AM, Zheng Y, Cancelas JA. Antioxidant prevents clearance of hemostatically competent platelets after long-term cold storage. Transfusion 2020; 61:557-567. [PMID: 33247486 DOI: 10.1111/trf.16200] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 09/27/2020] [Accepted: 10/23/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND Cold storage of platelets (PLTs) has the potential advantage of prolonging storage time while reducing posttransfusion infection given the decreased likelihood of bacterial outgrowth during storage and possibly beneficial effects in treating bleeding patients. However, cold storage reduces PLT survival through the induction of complex storage lesions, which are more accentuated when storage is prolonged. STUDY DESIGN AND METHODS Whole blood-derived PLT-rich plasma concentrates from seven PLT pools (n = 5 donors per pool). PLT additive solution was added (67%/33% plasma) and the product was split into 50-mL bags. Split units were stored in the presence or absence of 1 mM of N-acetylcysteine (NAC) under agitation for up to 14 days at room temperature or in the cold and were analyzed for PLT activation, fibrinogen-dependent spreading, microparticle formation, mitochondrial respiratory activity, reactive oxygen species (ROS) generation, as well as in vivo survival and bleeding time correction in immunodeficient mice. RESULTS Cold storage of PLTs for 7 days or longer induces significant PLT activation, cytoskeletal damage, impaired fibrinogen spreading, enhances mitochondrial metabolic decoupling and ROS generation, and increases macrophage-dependent phagocytosis and macrophage-independent clearance. Addition of NAC prevents PLT clearance and allows a correction of the prolonged bleeding time in thrombocytopenic, aspirin-treated, immunodeficient mice. CONCLUSIONS Long-term cold storage induces mitochondrial uncoupling and increased proton leak and ROS generation. The resulting ROS is a crucial contributor to the increased macrophage-dependent and -independent clearance of functional PLTs and can be prevented by the antioxidant NAC in a magnesium-containing additive solution.
Collapse
Affiliation(s)
- Shailaja Hegde
- Hoxworth Blood Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Ashley M Wellendorf
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Yi Zheng
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Jose A Cancelas
- Hoxworth Blood Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
57
|
Deppermann C, Kratofil RM, Peiseler M, David BA, Zindel J, Castanheira FVES, van der Wal F, Carestia A, Jenne CN, Marth JD, Kubes P. Macrophage galactose lectin is critical for Kupffer cells to clear aged platelets. J Exp Med 2020; 217:133651. [PMID: 31978220 PMCID: PMC7144524 DOI: 10.1084/jem.20190723] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 10/01/2019] [Accepted: 12/17/2019] [Indexed: 12/21/2022] Open
Abstract
Every day, megakaryocytes produce billions of platelets that circulate for several days and eventually are cleared by the liver. The exact removal mechanism, however, remains unclear. Loss of sialic acid residues is thought to feature in the aging and clearance of platelets. Using state-of-the-art spinning disk intravital microscopy to delineate the different compartments and cells of the mouse liver, we observed rapid accumulation of desialylated platelets predominantly on Kupffer cells, with only a few on endothelial cells and none on hepatocytes. Kupffer cell depletion prevented the removal of aged platelets from circulation. Ashwell-Morell receptor (AMR) deficiency alone had little effect on platelet uptake. Macrophage galactose lectin (MGL) together with AMR mediated clearance of desialylated or cold-stored platelets by Kupffer cells. Effective clearance is critical, as mice with an aged platelet population displayed a bleeding phenotype. Our data provide evidence that the MGL of Kupffer cells plays a significant role in the removal of desialylated platelets through a collaboration with the AMR, thereby maintaining a healthy and functional platelet compartment.
Collapse
Affiliation(s)
- Carsten Deppermann
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada.,Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Rachel M Kratofil
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Moritz Peiseler
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Bruna A David
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Joel Zindel
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Fernanda Vargas E Silva Castanheira
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Fardau van der Wal
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Agostina Carestia
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada.,Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Craig N Jenne
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada.,Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Jamey D Marth
- Center for Nanomedicine, SBP Medical Discovery Institute, and Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA
| | - Paul Kubes
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
58
|
Belizaire R, Makar RS. Non-Alloimmune Mechanisms of Thrombocytopenia and Refractoriness to Platelet Transfusion. Transfus Med Rev 2020; 34:242-249. [PMID: 33129606 PMCID: PMC7494440 DOI: 10.1016/j.tmrv.2020.09.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 08/24/2020] [Accepted: 09/03/2020] [Indexed: 12/11/2022]
Abstract
Refractoriness to platelet transfusion is a common clinical problem encountered by the transfusion medicine specialist. It is well recognized that most causes of refractoriness to platelet transfusion are not a consequence of alloimmunization to human leukocyte, platelet-specific, or ABO antigens, but are a consequence of platelet sequestration and consumption. This review summarizes the clinical factors that result in platelet refractoriness and highlights recent data describing novel biological mechanisms that contribute to this clinical problem.
Collapse
Affiliation(s)
- Roger Belizaire
- Associate Director, Adult Transfusion Medicine, Brigham and Women's Hospital, Boston, MA
| | - Robert S Makar
- Director, Blood Transfusion Service, Massachusetts General Hospital, Boston, MA.
| |
Collapse
|
59
|
Mack JP, Miles J, Stolla M. Cold-Stored Platelets: Review of Studies in Humans. Transfus Med Rev 2020; 34:221-226. [PMID: 33039263 DOI: 10.1016/j.tmrv.2020.08.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/17/2020] [Accepted: 08/20/2020] [Indexed: 01/12/2023]
Abstract
Although numerous reviews and editorials have been published about the biologic features of platelets exposed to cold temperature and their in vitro function, none has focused on the data from studies after transfusion in healthy human participants and patients. This may, in part, be due to the paucity of well-controlled in vivo investigations of cold-stored platelets. Although numerous studies are looking into the recovery and survival of cold-stored platelets (ie, the percentage of infused platelets maintained in circulation over time), very few assess in vivo platelet function. Another caveat is that most studies were performed in the 1960s and 1970s, at a time when platelet collection and storage were different compared to today. Despite these limitations, we believe the transfusion community can take valuable information from these studies. This review is limited to data on cold-stored platelets in plasma or additive solution and does not include data on whole blood or resuspended whole blood from components because the hemostatic properties of whole blood are likely very different (the interested reader is referred to the review article focused on the hemostatic properties of platelets stored in whole blood by van der Meer et al in this special edition of Transfusion Medicine Reviews). In this review, we report that room temperature storage consistently results in a longer in vivo platelet circulation time at the expense of bacterial growth and shorter storage duration, resulting in expiration, wastage, and regional and national shortages. Cold storage of platelets universally results in moderately reduced recovery and markedly reduced survival. We found inconsistent data about the efficacy of cold-stored platelets likely due to study design differences. The analysis of the available data suggests that there is a short-lasting hemostatic effect of cold-stored platelets. Storage time or choice of anticoagulant did not have a clear effect on platelet efficacy after cold storage. In summary, more data and clinical trials are needed to better understand the effect of cold-stored platelets after transfusion into humans.
Collapse
Affiliation(s)
- Johnathan P Mack
- University of Ottawa Medical Center, University of Ottawa, Ottawa, Ontario, Canada
| | - Jeffrey Miles
- Bloodworks Northwest Research Institute, Seattle, WA
| | - Moritz Stolla
- Bloodworks Northwest Research Institute, Seattle, WA; Department of Medicine, Division of Hematology, University of Washington School of Medicine, Seattle, WA.
| |
Collapse
|
60
|
Affiliation(s)
- Jack Yule
- Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham , Midlands, UK.,Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham , Edgbaston, UK
| | - Neil V Morgan
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham , Edgbaston, UK
| | - Natalie S Poulter
- Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham , Midlands, UK.,Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham , Edgbaston, UK
| |
Collapse
|
61
|
Lee-Sundlov MM, Stowell SR, Hoffmeister KM. Multifaceted role of glycosylation in transfusion medicine, platelets, and red blood cells. J Thromb Haemost 2020; 18:1535-1547. [PMID: 32350996 PMCID: PMC7336546 DOI: 10.1111/jth.14874] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 03/20/2020] [Accepted: 04/27/2020] [Indexed: 12/17/2022]
Abstract
Glycosylation is highly prevalent, and also one of the most complex and varied posttranslational modifications. This large glycan diversity results in a wide range of biological functions. Functional diversity includes protein degradation, protein clearance, cell trafficking, cell signaling, host-pathogen interactions, and immune defense, including both innate and acquired immunity. Glycan-based ABO(H) antigens are critical in providing compatible products in the setting of transfusion and organ transplantation. However, evidence also suggests that ABO expression may influence cardiovascular disease, thrombosis, and hemostasis disorders, including alterations in platelet function and von Willebrand factor blood levels. Glycans also regulate immune and hemostasis function beyond ABO(H) antigens. Mutations in glycogenes (PIGA, COSMC) lead to serious blood disorders, including Tn syndrome associated with hyperagglutination, hemolysis, and thrombocytopenia. Alterations in genes responsible for sialic acids (Sia) synthesis (GNE) and UDP-galactose (GALE) and lactosamine (LacNAc) (B4GALT1) profoundly affect circulating platelet counts. Desialylation (removal of Sia) is affected by human and pathogenic neuraminidases. This review addresses the role of glycans in transfusion medicine, hemostasis and thrombosis, and red blood cell and platelet survival.
Collapse
Affiliation(s)
- Melissa M. Lee-Sundlov
- Translational Glycomics Center, Blood Research Institute Versiti, Milwaukee, WI, United States
| | - Sean R. Stowell
- Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology, Emory University School of Medicine, Atlanta, GA, United States
| | - Karin M. Hoffmeister
- Translational Glycomics Center, Blood Research Institute Versiti, Milwaukee, WI, United States
- Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology, Emory University School of Medicine, Atlanta, GA, United States
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee WI, United States
| |
Collapse
|
62
|
Lasne D, Pascreau T, Darame S, Bourrienne M, Tournoux P, Philippe A, Ziachahabi S, Suarez F, Marcais A, Dupont A, Denis CV, Kauskot A, Borgel D. Measuring beta-galactose exposure on platelets: Standardization and healthy reference values. Res Pract Thromb Haemost 2020; 4:813-822. [PMID: 33134771 PMCID: PMC7586713 DOI: 10.1002/rth2.12369] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 04/28/2020] [Accepted: 04/30/2020] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Correct diagnosis of the cause of thrombocytopenia is crucial for the appropriate management of patients. Hyposialylation/desialylation (characterized by abnormally high β-galactose exposure) accelerates platelet clearance and can lead to thrombocytopenia. However, the reference range for β-galactose exposure in healthy individuals has not been defined previously. OBJECTIVE The objective of the present study was to develop a standardized assay of platelet β-galactose exposure for implementation in a clinical laboratory. METHODS β-Galactose exposure was measured in platelet-rich plasma by using flow cytometry and Ricinus communis agglutinin (RCA). A population of 120 healthy adults was recruited to study variability. RESULTS We determined an optimal RCA concentration of 12.5 μg/mL. The measure was stable for up to 4 hours (mean fluorescence intensity [MFI]-RCA: 1233 ± 329 at 0 hour and 1480 ± 410 at 4 hours). The platelet count did not induce a variation of RCA and the measure of RCA was stable when tested up to 24 hours after blood collection (MFI-RCA: 1252 ± 434 at day 0 and 1140 ± 297 24 hours after blood sampling). To take into account the platelet size, results should be expressed as RCA/forward scatter ratio. We used the assay to study variability in 120 healthy adults, and we found that the ratio is independent of sex and blood group. CONCLUSION We defined a normal range in a healthy population and several preanalytical and analytical variables were evaluated, together with positive and negative controls. This assay may assist in the diagnosis of thrombocytopenic diseases linked to changes in β-galactose exposure.
Collapse
Affiliation(s)
- Dominique Lasne
- Department of Biological HematologyHôpital NeckerAP‐HPParisFrance
- HIThUMR_S 1176INSERMUniv. Paris‐SaclayLe Kremlin‐BicêtreFrance
| | - Tiffany Pascreau
- Department of Biological HematologyHôpital NeckerAP‐HPParisFrance
- HIThUMR_S 1176INSERMUniv. Paris‐SaclayLe Kremlin‐BicêtreFrance
| | - Sadyo Darame
- Department of Biological HematologyHôpital NeckerAP‐HPParisFrance
| | | | - Peggy Tournoux
- Department of Biological HematologyHôpital NeckerAP‐HPParisFrance
| | | | - Sara Ziachahabi
- Department of Biological HematologyHôpital NeckerAP‐HPParisFrance
| | - Felipe Suarez
- Department of HematologyHôpital NeckerAP‐HPParisFrance
| | | | - Annabelle Dupont
- Department of Haemostasis and TransfusionCHU LilleLilleFrance
- Institut Pasteur de LilleU1011‐ EGIDInsermCHU LilleUniv. LilleLilleFrance
| | - Cécile V. Denis
- HIThUMR_S 1176INSERMUniv. Paris‐SaclayLe Kremlin‐BicêtreFrance
| | | | - Delphine Borgel
- Department of Biological HematologyHôpital NeckerAP‐HPParisFrance
- HIThUMR_S 1176INSERMUniv. Paris‐SaclayLe Kremlin‐BicêtreFrance
| |
Collapse
|
63
|
Papa AL, Jiang A, Korin N, Chen MB, Langan ET, Waterhouse A, Nash E, Caroff J, Graveline A, Vernet A, Mammoto A, Mammoto T, Jain A, Kamm RD, Gounis MJ, Ingber DE. Platelet decoys inhibit thrombosis and prevent metastatic tumor formation in preclinical models. Sci Transl Med 2020; 11:11/479/eaau5898. [PMID: 30760580 DOI: 10.1126/scitranslmed.aau5898] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 06/26/2018] [Accepted: 01/19/2019] [Indexed: 01/05/2023]
Abstract
Platelets are crucial for normal hemostasis; however, their hyperactivation also contributes to many potentially lethal pathologies including myocardial infarction, stroke, and cancer. We hypothesized that modified platelets lacking their aggregation and activation capacity could act as reversible inhibitors of platelet activation cascades. Here, we describe the development of detergent-extracted human modified platelets (platelet decoys) that retained platelet binding functions but were incapable of functional activation and aggregation. Platelet decoys inhibited aggregation and adhesion of platelets on thrombogenic surfaces in vitro, which could be immediately reversed by the addition of normal platelets; in vivo in a rabbit model, pretreatment with platelet decoys inhibited arterial injury-induced thromboembolism. Decoys also interfered with platelet-mediated human breast cancer cell aggregation, and their presence decreased cancer cell arrest and extravasation in a microfluidic human microvasculature on a chip. In a mouse model of metastasis, simultaneous injection of the platelet decoys with tumor cells inhibited metastatic tumor growth. Thus, our results suggest that platelet decoys might represent an effective strategy for obtaining antithrombotic and antimetastatic effects.
Collapse
Affiliation(s)
- Anne-Laure Papa
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA. .,Department of Biomedical Engineering, The George Washington University, Washington, DC 20052, USA
| | - Amanda Jiang
- Vascular Biology Program and Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Netanel Korin
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Michelle B Chen
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Erin T Langan
- New England Center for Stroke Research, Department of Radiology, University of Massachusetts, Worcester, MA 01655, USA
| | - Anna Waterhouse
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Emma Nash
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Jildaz Caroff
- New England Center for Stroke Research, Department of Radiology, University of Massachusetts, Worcester, MA 01655, USA
| | - Amanda Graveline
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Andyna Vernet
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Akiko Mammoto
- Vascular Biology Program and Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Tadanori Mammoto
- Vascular Biology Program and Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Abhishek Jain
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Roger D Kamm
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Matthew J Gounis
- New England Center for Stroke Research, Department of Radiology, University of Massachusetts, Worcester, MA 01655, USA
| | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA. .,Vascular Biology Program and Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.,Harvard John A. Paulson School of Engineering and Applied Sciences, Cambridge, MA 02138, USA
| |
Collapse
|
64
|
Li JL, Zarbock A, Hidalgo A. Platelets as autonomous drones for hemostatic and immune surveillance. J Exp Med 2020; 214:2193-2204. [PMID: 28720569 PMCID: PMC5551582 DOI: 10.1084/jem.20170879] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 06/26/2017] [Accepted: 07/06/2017] [Indexed: 12/13/2022] Open
Abstract
Platelets participate in many important physiological processes, including hemostasis and immunity. However, despite their broad participation in these evolutionarily critical roles, the anucleate platelet is uniquely mammalian. In contrast with the large nucleated equivalents in lower vertebrates, we find that the design template for the evolutionary specialization of platelets shares remarkable similarities with human-engineered unmanned aerial vehicles in terms of overall autonomy, maneuverability, and expendability. Here, we review evidence illustrating how platelets are uniquely suited for surveillance and the manner in which they consequently provide various types of support to other cell types.
Collapse
Affiliation(s)
- Jackson LiangYao Li
- Area of Developmental and Cell Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain.,Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, Singapore
| | - Alexander Zarbock
- Department of Anesthesiology, Intensive Care, and Pain Medicine, University of Münster, Münster, Germany
| | - Andrés Hidalgo
- Area of Developmental and Cell Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain.,Institute for Cardiovascular Prevention, Ludwig-Maximillians-University, Munich, Germany
| |
Collapse
|
65
|
β4GALT1 controls β1 integrin function to govern thrombopoiesis and hematopoietic stem cell homeostasis. Nat Commun 2020; 11:356. [PMID: 31953383 PMCID: PMC6968998 DOI: 10.1038/s41467-019-14178-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 12/13/2019] [Indexed: 12/26/2022] Open
Abstract
Glycosylation is critical to megakaryocyte (MK) and thrombopoiesis in the context of gene mutations that affect sialylation and galactosylation. Here, we identify the conserved B4galt1 gene as a critical regulator of thrombopoiesis in MKs. β4GalT1 deficiency increases the number of fully differentiated MKs. However, the resulting lack of glycosylation enhances β1 integrin signaling leading to dysplastic MKs with severely impaired demarcation system formation and thrombopoiesis. Platelets lacking β4GalT1 adhere avidly to β1 integrin ligands laminin, fibronectin, and collagen, while other platelet functions are normal. Impaired thrombopoiesis leads to increased plasma thrombopoietin (TPO) levels and perturbed hematopoietic stem cells (HSCs). Remarkably, β1 integrin deletion, specifically in MKs, restores thrombopoiesis. TPO and CXCL12 regulate β4GalT1 in the MK lineage. Thus, our findings establish a non-redundant role for β4GalT1 in the regulation of β1 integrin function and signaling during thrombopoiesis. Defective thrombopoiesis and lack of β4GalT1 further affect HSC homeostasis.
Collapse
|
66
|
Yu Z, Shibazaki M, Otsuka H, Takada H, Nakamura M, Endo Y. Dynamics of Platelet Behaviors as Defenders and Guardians: Accumulations in Liver, Lung, and Spleen in Mice. Biol Pharm Bull 2020; 42:1253-1267. [PMID: 31366863 DOI: 10.1248/bpb.b18-00975] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Systemic platelet behaviors in experimental animals are often assessed by infusion of isotope-labeled platelets and measuring them under anesthesia. However, such procedures alter, therefore may not reveal, real-life platelet behaviors. 5-Hydroxytryptamine (5HT or serotonin) is present within limited cell-types, including platelets. In our studies, by measuring 5HT as a platelet-marker in non-anesthetized mice, we identified stimulation- and time-dependent accumulations in liver, lung, and/or spleen as important systemic platelet behaviors. For example, intravenous, intraperitoneal, or intragingival injection of lipopolysaccharide (LPS, a cell-wall component of Gram-negative bacteria), interleukin (IL)-1, or tumor necrosis factor (TNF)-α induced hepatic platelet accumulation (HPA) and platelet translocation into the sinusoidal and perisinusoidal spaces or hepatocytes themselves. These events occurred "within a few hours" of the injection, caused hypoglycemia, and exhibited protective or causal effects on hepatitis. Intravenous injection of larger doses of LPS into normal mice, or intravenous antigen-challenge to sensitized mice, induced pulmonary platelet accumulation (PPA), as well as HPA. These reactions occurred "within a few min" of the LPS injection or antigen challenge and resulted in shock. Intravenous injection of 5HT or a catecholamine induced a rapid PPA "within 6 s." Intravenous LPS injection, within a minute, increased the pulmonary catecholamines that mediate the LPS-induced PPA. Macrophage-depletion from liver and spleen induced "day-scale" splenic platelet accumulation, suggesting the spleen is involved in clearing senescent platelets. These findings indicate the usefulness of 5HT as a marker of platelet behaviors, and provide a basis for a discussion of the roles of platelets as both "defenders" and "guardians."
Collapse
Affiliation(s)
- Zhiqian Yu
- Department of Disaster Psychiatry, International Research Institute for Disaster Science, Tohoku University
| | - Masahiro Shibazaki
- Department of Tumor Biology, Institute of Biomedical Sciences, Iwate Medical University
| | - Hirotada Otsuka
- Laboratory of Veterinary Anatomy, School of Veterinary Medicine, Faculty of Veterinary Science, Nippon Veterinary and Life Science University
| | - Haruhiko Takada
- Department of Microbiology and Immunology, Graduate School of Dentistry, Tohoku University
| | - Masanori Nakamura
- Department of Oral Anatomy and Developmental Biology, School of Dentistry, Showa University
| | - Yasuo Endo
- Division of Oral and Maxillofacial Surgery, Graduate School of Dentistry, Tohoku University
| |
Collapse
|
67
|
Lu M, Dai T, Hu S, Zhang Q, Bhayana B, Wang L, Wu MX. Antimicrobial blue light for decontamination of platelets during storage. JOURNAL OF BIOPHOTONICS 2020; 13:e201960021. [PMID: 31407467 PMCID: PMC7083650 DOI: 10.1002/jbio.201960021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 07/31/2019] [Accepted: 08/07/2019] [Indexed: 05/28/2023]
Abstract
Platelet (PLT) storage is currently limited to 5 days in clinics in the United States, in part, due to an increasing risk for microbial contamination over time. In light of well-documented antimicrobial activity of blue light (405-470 nm), we investigated potentials to decontaminate microbes during PLT storage by antimicrobial blue light (aBL). We found that PLTs produced no detectable levels of porphyrins or their derivatives, the chromophores that specifically absorb blue light, in marked contrast to microbes that generated porphyrins abundantly. The difference formed a basis with which aBL selectively inactivated contaminated microbes prior to and during the storage, without incurring any harm to PLTs. In accordance with this, when contamination with representative microbes was simulated in PLT concentrates supplemented with 65% of PLT additive solution in a standard storage bag, all "contaminated" microbes tested were completely inactivated after exposure of the bag to 405 nm aBL at 75 J/cm2 only once. While killing microbes efficiently, this dose of aBL irradiation exerted no adverse effects on the viability, activation or aggregation of PLTs ex vivo and could be used repeatedly during PLT storage. PLT survival in vivo was also unaltered by aBL irradiation after infusion of aBL-irradiated mouse PLTs into mice. The study provides proof-of-concept evidence for a potential of aBL to decontaminate PLTs during storage.
Collapse
Affiliation(s)
- Min Lu
- Department of Dermatology, Harvard Medical School, Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts
| | - TianHong Dai
- Department of Dermatology, Harvard Medical School, Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts
| | - SiSi Hu
- Department of Dermatology, Harvard Medical School, Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Qi Zhang
- Department of Dermatology, Harvard Medical School, Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Brijesh Bhayana
- Department of Dermatology, Harvard Medical School, Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Li Wang
- Department of Dermatology, Harvard Medical School, Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Mei X. Wu
- Department of Dermatology, Harvard Medical School, Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts
| |
Collapse
|
68
|
Baghdadi V, Yari F, Nikougoftar M, Rafiee MH. Platelets Apoptosis and Clearance in The Presence of Sodium Octanoate during Storage of Platelet Concentrate at 4˚C. CELL JOURNAL 2019; 22:212-217. [PMID: 31721536 PMCID: PMC6874783 DOI: 10.22074/cellj.2020.6697] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 03/09/2019] [Indexed: 12/19/2022]
Abstract
Objective Platelet (PLT) storage at 4˚C has several benefits, however, it is accompanied by increased clearance of
PLTs after transfusion. In this study, we evaluated the potential of sodium octanoate (SO) for reducing apoptosis and
clearance rate of PLTs after long-term storage in cold.
Materials and Methods In this experimental study, PLT concentrates (PCs) were stored for 5 days under the following
three conditions: 20-24˚C, 4˚C, and 4˚C in the presence of SO. To measure the viability of PLTs, the water-soluble
tetrazolium salt (WST-1) assay was performed. Phosphatidylserine (PS) exposure was determined on PLTs using
flow cytometry technique. The amount of human active caspase-3 was determined in PLTs using an enzyme-linked
immunosorbent assay. Additionally, the amount of PLT ingestion or clearance was determined by using HepG2 cell line.
Results The viability was higher in the SO-treated PLTs compared to the other groups. The level of PS exposure
on PLTs was lower in the SO-treated PLTs compared to the other groups. The amount of active caspase-3 increased
in all groups during 5-day storage. The highest increase in the amount of caspase-3 levels was observed at cold
temperature. However, PLTs kept at 4˚C in the presence of SO had a lower amount of active caspase-3 compared to
PLTs kept at 4˚C. The amount of PLTs removal by HepG2 cells was increased for 4˚C-kept PLTs but it was lower for
PLTs kept at 4˚C in the presence of SO but, the differences were not significant (P>0.05).
Conclusion SO could partially moderate the effects of cold temperature on apoptosis and viability of platelets. It also
decreases the ingestion rate of long-time refrigerated PLTs in vitro. Further studies using higher numbers of samples
are required to demonstrate the effect of SO on reducing the clearance rate of PLTs.
Collapse
Affiliation(s)
- Vahid Baghdadi
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Fatemeh Yari
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran. Elevtronic Address:
| | - Mahin Nikougoftar
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Mohammad Hessam Rafiee
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| |
Collapse
|
69
|
Chen 陈温纯 W, Voos KM, Josephson CD, Li R. Short-Acting Anti-VWF (von Willebrand Factor) Aptamer Improves the Recovery, Survival, and Hemostatic Functions of Refrigerated Platelets. Arterioscler Thromb Vasc Biol 2019; 39:2028-2037. [PMID: 31315441 DOI: 10.1161/atvbaha.119.312439] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Refrigeration-induced binding of VWF (von Willebrand factor) to platelets contributes to the rapid clearance of refrigerated platelets. In this study, we investigate whether inhibiting VWF binding by a DNA-based aptamer ameliorates the clearance of refrigerated platelets without significantly impeding hemostatic functions. Approach and Results: Platelets were refrigerated with or without aptamer ARC1779 for 48 hours. VWF binding, the effective lifetime of ARC1779, platelet post-transfusion recovery and survival, and the hemostatic function were measured. ARC1779 treatment during refrigeration inhibited the platelet-VWF interaction. ARC1779-treated refrigerated murine platelets exhibited increased post-transfusion recovery and survival than untreated ones (recovery of ARC1779-treated platelets: 76.7±5.5%; untreated: 63.7±0.8%; P<0.01. Half-life: 31.4±2.36 hours versus 28.1±0.86 hours; P<0.05). A similar increase was observed for refrigerated human platelets (recovery: 49.4±4.4% versus 36.8±2.1%, P<0.01; half-life: 9.2±1.5 hours versus 8.7±0.9 hours, ns). The effective lifetime of ARC1779 in mice was 2 hours. Additionally, ARC1779 improved the long-term (2 hours after transfusion) hemostatic function of refrigerated platelets (tail bleeding time of mice transfused with ARC1779-treated refrigerated platelets: 160±65 seconds; untreated: 373±96 seconds; P<0.01). The addition of an ARC1779 antidote before transfusion improved the immediate (15 minutes after transfusion) hemostatic function (bleeding time of treated platelets: 149±21 seconds; untreated: 320±36 seconds; P<0.01). CONCLUSIONS ARC1779 improves the post-transfusion recovery of refrigerated platelets and preserves the long-term hemostatic function of refrigerated platelets. These results suggest that a short-acting inhibitor of the platelet-VWF interaction may be a potential therapeutic option to improve refrigeration of platelets for transfusion treatment.
Collapse
Affiliation(s)
- Wenchun Chen 陈温纯
- From the Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Department of Pediatrics (W.C., K.M.V., C.D.J., R.L.), Emory University School of Medicine, Atlanta, GA
| | - Kayleigh M Voos
- From the Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Department of Pediatrics (W.C., K.M.V., C.D.J., R.L.), Emory University School of Medicine, Atlanta, GA
| | - Cassandra D Josephson
- From the Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Department of Pediatrics (W.C., K.M.V., C.D.J., R.L.), Emory University School of Medicine, Atlanta, GA.,Department of Pathology (C.D.J), Emory University School of Medicine, Atlanta, GA
| | - Renhao Li
- From the Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Department of Pediatrics (W.C., K.M.V., C.D.J., R.L.), Emory University School of Medicine, Atlanta, GA
| |
Collapse
|
70
|
|
71
|
Six KR, Devloo R, Compernolle V, Feys HB. Impact of cold storage on platelets treated with Intercept pathogen inactivation. Transfusion 2019; 59:2662-2671. [PMID: 31187889 PMCID: PMC6851707 DOI: 10.1111/trf.15398] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 05/23/2019] [Accepted: 05/23/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND Pathogen inactivation and cold or cryopreservation of platelets (PLTs) both significantly affect PLT function. It is not known how PLTs function when both are combined. STUDY DESIGN AND METHODS Standard PLT concentrates (PCs) were compared to pathogen‐inactivated PCs treated with amotosalen photochemical treatment (AS‐PCT) when stored at room (RT, 22°C), cold (4°C, n = 6), or cryopreservation (−80°C, n = 8) temperatures. The impact of alternative storage methods on both arms was studied in flow cytometry, light transmittance aggregometry, and hemostasis in collagen‐coated microfluidic flow chambers. RESULTS Platelet aggregation of cold‐stored AS‐PCT PLTs was 44% ± 11% compared to 57% ± 14% for cold‐stored standard PLTs and 58% ± 21% for RT‐stored AS‐PCT PLTs. Integrin activation of cold‐stored AS‐PCT PLTs was 53% ± 9% compared to 77% ± 6% for cold‐stored standard PLTs and 69% ± 13% for RT‐stored AS‐PCT PLTs. Coagulation of cold‐stored AS‐PCT PLTs started faster under flow (836 ± 140 sec) compared to cold‐stored standard PLTs (960 ± 192 sec) and RT‐stored AS‐PCT PLTs (1134 ± 220 sec). Fibrin formation rate under flow was also highest for cold‐stored AS‐PCT PLTs. This was in line with thrombin generation in static conditions because cold‐stored AS‐PCT PLTs generated 297 ± 47 nmol/L thrombin compared to 159 ± 33 nmol/L for cold‐stored standard PLTs and 83 ± 25 nmol/L for RT‐stored AS‐PCT PLTs. So despite decreased PLT activation and aggregation, cold storage of AS‐PCT PLTs promoted coagulation. PLT aggregation of cryopreserved AS‐PCT PLTs (23% ± 10%) was not significantly different from cryopreserved standard PLTs (25% ± 8%). CONCLUSION This study shows that cold storage of AS‐PCT PLTs further affects PLT activation and aggregation but promotes (pro)coagulation. Increased procoagulation was not observed after cryopreservation.
Collapse
Affiliation(s)
- Katrijn R Six
- Transfusion Research Center, Belgian Red Cross-Flanders, Ghent, Belgium.,Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Rosalie Devloo
- Transfusion Research Center, Belgian Red Cross-Flanders, Ghent, Belgium
| | - Veerle Compernolle
- Transfusion Research Center, Belgian Red Cross-Flanders, Ghent, Belgium.,Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium.,Blood Service of the Belgian Red Cross-Flanders, Mechelen, Belgium
| | - Hendrik B Feys
- Transfusion Research Center, Belgian Red Cross-Flanders, Ghent, Belgium.,Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| |
Collapse
|
72
|
Toonstra C, Hu Y, Zhang H. Deciphering the Roles of N-Glycans on Collagen-Platelet Interactions. J Proteome Res 2019; 18:2467-2477. [PMID: 31055923 DOI: 10.1021/acs.jproteome.9b00003] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Collagen is a potent agonist for platelet activation, presenting itself as a key contributor to coagulation via interactions with platelet glycoproteins. The fine details dictating platelet-collagen interactions are poorly understood. In particular, glycosylation could be a key determinant in the platelet-collagen interaction. Here, we report an affinity purification coupled to a mass spectrometry-based approach to elucidate the function of N-glycans in dictating platelet-collagen interactions. By integrative proteomic and glycoproteomic analysis of collagen-platelet interactive proteins with N-glycan manipulation, we demonstrate that the interaction of platelet adhesive receptors with collagen is highly N-glycan regulated, with glycans on many receptors playing positive roles in collagen binding, with glycans on other platelet glycoproteins exhibiting inhibitory roles on the binding to collagen. Our results significantly enhance our understanding of the details of glycans influencing the platelet-collagen interaction.
Collapse
Affiliation(s)
- Christian Toonstra
- Department of Pathology , Johns Hopkins School of Medicine , 400 N Broadway , Baltimore , Maryland 21287 , United States
| | - Yingwei Hu
- Department of Pathology , Johns Hopkins School of Medicine , 400 N Broadway , Baltimore , Maryland 21287 , United States
| | - Hui Zhang
- Department of Pathology , Johns Hopkins School of Medicine , 400 N Broadway , Baltimore , Maryland 21287 , United States
| |
Collapse
|
73
|
Dupont A, Soukaseum C, Cheptou M, Adam F, Nipoti T, Lourenco-Rodrigues MD, Legendre P, Proulle V, Rauch A, Kawecki C, Bryckaert M, Rosa JP, Paris C, Ternisien C, Boisseau P, Goudemand J, Borgel D, Lasne D, Maurice P, Lenting PJ, Denis CV, Susen S, Kauskot A. Relevance of platelet desialylation and thrombocytopenia in type 2B von Willebrand disease: preclinical and clinical evidence. Haematologica 2019; 104:2493-2500. [PMID: 30819911 PMCID: PMC6959185 DOI: 10.3324/haematol.2018.206250] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 02/26/2019] [Indexed: 11/13/2022] Open
Abstract
Patients with type 2B von Willebrand disease (vWD) (caused by gain-of-function mutations in the gene coding for von Willebrand factor) display bleeding to a variable extent and, in some cases, thrombocytopenia. There are several underlying causes of thrombocytopenia in type 2B vWD. It was recently suggested that desialylation-mediated platelet clearance leads to thrombocytopenia in this disease. However, this hypothesis has not been tested in vivo. The relationship between platelet desialylation and the platelet count was probed in 36 patients with type 2B von Willebrand disease (p.R1306Q, p.R1341Q, and p.V1316M mutations) and in a mouse model carrying the severe p.V1316M mutation (the 2B mouse). We observed abnormally high elevated levels of platelet desialylation in both patients with the p.V1316M mutation and the 2B mice. In vitro, we demonstrated that 2B p.V1316M/von Willebrand factor induced more desialylation of normal platelets than wild-type von Willebrand factor did. Furthermore, we found that N-glycans were desialylated and we identified αIIb and β3 as desialylation targets. Treatment of 2B mice with sialidase inhibitors (which correct platelet desialylation) was not associated with the recovery of a normal platelet count. Lastly, we demonstrated that a critical platelet desialylation threshold (not achieved in either 2B patients or 2B mice) was required to induce thrombocytopenia in vivo. In conclusion, in type 2B vWD, platelet desialylation has a minor role and is not sufficient to mediate thrombocytopenia.
Collapse
Affiliation(s)
- Annabelle Dupont
- Université de Lille, UMR Inserm 1011, Institut Pasteur de Lille, EGID, F-59000 Lille.,Department of Hematology, CHU de Lille, F-59000 Lille
| | - Christelle Soukaseum
- HITh, UMR_S 1176, INSERM Université Paris-Sud, Université Paris-Saclay, F-94270 Le Kremlin-Bicêtre
| | - Mathilde Cheptou
- HITh, UMR_S 1176, INSERM Université Paris-Sud, Université Paris-Saclay, F-94270 Le Kremlin-Bicêtre
| | - Frédéric Adam
- HITh, UMR_S 1176, INSERM Université Paris-Sud, Université Paris-Saclay, F-94270 Le Kremlin-Bicêtre
| | - Thomas Nipoti
- HITh, UMR_S 1176, INSERM Université Paris-Sud, Université Paris-Saclay, F-94270 Le Kremlin-Bicêtre
| | | | - Paulette Legendre
- HITh, UMR_S 1176, INSERM Université Paris-Sud, Université Paris-Saclay, F-94270 Le Kremlin-Bicêtre
| | - Valérie Proulle
- HITh, UMR_S 1176, INSERM Université Paris-Sud, Université Paris-Saclay, F-94270 Le Kremlin-Bicêtre.,AP-HP, Department of Biological Hematology, CHU Bicêtre, Hôpitaux Universitaires Paris Sud, F-94270 Paris
| | - Antoine Rauch
- Université de Lille, UMR Inserm 1011, Institut Pasteur de Lille, EGID, F-59000 Lille.,Department of Hematology, CHU de Lille, F-59000 Lille
| | - Charlotte Kawecki
- HITh, UMR_S 1176, INSERM Université Paris-Sud, Université Paris-Saclay, F-94270 Le Kremlin-Bicêtre
| | - Marijke Bryckaert
- HITh, UMR_S 1176, INSERM Université Paris-Sud, Université Paris-Saclay, F-94270 Le Kremlin-Bicêtre
| | - Jean-Philippe Rosa
- HITh, UMR_S 1176, INSERM Université Paris-Sud, Université Paris-Saclay, F-94270 Le Kremlin-Bicêtre
| | - Camille Paris
- Department of Hematology, CHU de Lille, F-59000 Lille
| | | | | | - Jenny Goudemand
- Université de Lille, UMR Inserm 1011, Institut Pasteur de Lille, EGID, F-59000 Lille.,Department of Hematology, CHU de Lille, F-59000 Lille
| | - Delphine Borgel
- HITh, UMR_S 1176, INSERM Université Paris-Sud, Université Paris-Saclay, F-94270 Le Kremlin-Bicêtre.,AP-HP, Department of Biological Hematology, Hôpital Necker, F-75015 Paris
| | - Dominique Lasne
- HITh, UMR_S 1176, INSERM Université Paris-Sud, Université Paris-Saclay, F-94270 Le Kremlin-Bicêtre.,AP-HP, Department of Biological Hematology, Hôpital Necker, F-75015 Paris
| | - Pascal Maurice
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Team 2 "Matrix aging and Vascular remodelling", Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, F-51000 Reims, France
| | - Peter J Lenting
- HITh, UMR_S 1176, INSERM Université Paris-Sud, Université Paris-Saclay, F-94270 Le Kremlin-Bicêtre
| | - Cécile V Denis
- HITh, UMR_S 1176, INSERM Université Paris-Sud, Université Paris-Saclay, F-94270 Le Kremlin-Bicêtre
| | - Sophie Susen
- Université de Lille, UMR Inserm 1011, Institut Pasteur de Lille, EGID, F-59000 Lille.,Department of Hematology, CHU de Lille, F-59000 Lille
| | - Alexandre Kauskot
- HITh, UMR_S 1176, INSERM Université Paris-Sud, Université Paris-Saclay, F-94270 Le Kremlin-Bicêtre
| |
Collapse
|
74
|
|
75
|
Siddon AJ, Tormey CA, Snyder EL. Platelet Transfusion Medicine. Platelets 2019. [DOI: 10.1016/b978-0-12-813456-6.00064-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
76
|
|
77
|
Chen M, Yan R, Zhou K, Li X, Zhang Y, Liu C, Jiang M, Ye H, Meng X, Pang N, Zhao L, Liu J, Xiao W, Hu R, Cui Q, Zhong W, Zhao Y, Zhu M, Lin A, Ruan C, Dai K. Akt-mediated platelet apoptosis and its therapeutic implications in immune thrombocytopenia. Proc Natl Acad Sci U S A 2018; 115:E10682-E10691. [PMID: 30337485 PMCID: PMC6233141 DOI: 10.1073/pnas.1808217115] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Immune thrombocytopenia (ITP) is an autoimmune disorder characterized by low platelet count which can cause fatal hemorrhage. ITP patients with antiplatelet glycoprotein (GP) Ib-IX autoantibodies appear refractory to conventional treatments, and the mechanism remains elusive. Here we show that the platelets undergo apoptosis in ITP patients with anti-GPIbα autoantibodies. Consistent with these findings, the anti-GPIbα monoclonal antibodies AN51 and SZ2 induce platelet apoptosis in vitro. We demonstrate that anti-GPIbα antibody binding activates Akt, which elicits platelet apoptosis through activation of phosphodiesterase (PDE3A) and PDE3A-mediated PKA inhibition. Genetic ablation or chemical inhibition of Akt or blocking of Akt signaling abolishes anti-GPIbα antibody-induced platelet apoptosis. We further demonstrate that the antibody-bound platelets are removed in vivo through an apoptosis-dependent manner. Phosphatidylserine (PS) exposure on apoptotic platelets results in phagocytosis of platelets by macrophages in the liver. Notably, inhibition or genetic ablation of Akt or Akt-regulated apoptotic signaling or blockage of PS exposure protects the platelets from clearance. Therefore, our findings reveal pathogenic mechanisms of ITP with anti-GPIbα autoantibodies and, more importantly, suggest therapeutic strategies for thrombocytopenia caused by autoantibodies or other pathogenic factors.
Collapse
Affiliation(s)
- Mengxing Chen
- Jiangsu Institute of Hematology, The First Affiliated Hospital and Collaborative Innovation Center of Hematology, State Key Laboraotry of Radiation Medicine and Protection, Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Suzhou, Jiangsu 215006, China
| | - Rong Yan
- Jiangsu Institute of Hematology, The First Affiliated Hospital and Collaborative Innovation Center of Hematology, State Key Laboraotry of Radiation Medicine and Protection, Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Suzhou, Jiangsu 215006, China;
| | - Kangxi Zhou
- Jiangsu Institute of Hematology, The First Affiliated Hospital and Collaborative Innovation Center of Hematology, State Key Laboraotry of Radiation Medicine and Protection, Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Suzhou, Jiangsu 215006, China
| | - Xiaodong Li
- Jiangsu Institute of Hematology, The First Affiliated Hospital and Collaborative Innovation Center of Hematology, State Key Laboraotry of Radiation Medicine and Protection, Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Suzhou, Jiangsu 215006, China
| | - Yang Zhang
- Jiangsu Institute of Hematology, The First Affiliated Hospital and Collaborative Innovation Center of Hematology, State Key Laboraotry of Radiation Medicine and Protection, Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Suzhou, Jiangsu 215006, China
| | - Chunliang Liu
- Jiangsu Institute of Hematology, The First Affiliated Hospital and Collaborative Innovation Center of Hematology, State Key Laboraotry of Radiation Medicine and Protection, Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Suzhou, Jiangsu 215006, China
| | - Mengxiao Jiang
- Jiangsu Institute of Hematology, The First Affiliated Hospital and Collaborative Innovation Center of Hematology, State Key Laboraotry of Radiation Medicine and Protection, Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Suzhou, Jiangsu 215006, China
| | - Honglei Ye
- Jiangsu Institute of Hematology, The First Affiliated Hospital and Collaborative Innovation Center of Hematology, State Key Laboraotry of Radiation Medicine and Protection, Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Suzhou, Jiangsu 215006, China
| | - Xingjun Meng
- Jiangsu Institute of Hematology, The First Affiliated Hospital and Collaborative Innovation Center of Hematology, State Key Laboraotry of Radiation Medicine and Protection, Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Suzhou, Jiangsu 215006, China
| | - Ningbo Pang
- Jiangsu Institute of Hematology, The First Affiliated Hospital and Collaborative Innovation Center of Hematology, State Key Laboraotry of Radiation Medicine and Protection, Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Suzhou, Jiangsu 215006, China
| | - Lili Zhao
- Jiangsu Institute of Hematology, The First Affiliated Hospital and Collaborative Innovation Center of Hematology, State Key Laboraotry of Radiation Medicine and Protection, Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Suzhou, Jiangsu 215006, China
| | - Jun Liu
- Jiangsu Institute of Hematology, The First Affiliated Hospital and Collaborative Innovation Center of Hematology, State Key Laboraotry of Radiation Medicine and Protection, Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Suzhou, Jiangsu 215006, China
| | - Weiling Xiao
- Jiangsu Institute of Hematology, The First Affiliated Hospital and Collaborative Innovation Center of Hematology, State Key Laboraotry of Radiation Medicine and Protection, Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Suzhou, Jiangsu 215006, China
| | - Renping Hu
- Jiangsu Institute of Hematology, The First Affiliated Hospital and Collaborative Innovation Center of Hematology, State Key Laboraotry of Radiation Medicine and Protection, Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Suzhou, Jiangsu 215006, China
| | - Qingya Cui
- Jiangsu Institute of Hematology, The First Affiliated Hospital and Collaborative Innovation Center of Hematology, State Key Laboraotry of Radiation Medicine and Protection, Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Suzhou, Jiangsu 215006, China
| | - Wei Zhong
- Jiangsu Institute of Hematology, The First Affiliated Hospital and Collaborative Innovation Center of Hematology, State Key Laboraotry of Radiation Medicine and Protection, Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Suzhou, Jiangsu 215006, China
| | - Yunxiao Zhao
- Jiangsu Institute of Hematology, The First Affiliated Hospital and Collaborative Innovation Center of Hematology, State Key Laboraotry of Radiation Medicine and Protection, Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Suzhou, Jiangsu 215006, China
| | - Mingqing Zhu
- Jiangsu Institute of Hematology, The First Affiliated Hospital and Collaborative Innovation Center of Hematology, State Key Laboraotry of Radiation Medicine and Protection, Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Suzhou, Jiangsu 215006, China
| | - Anning Lin
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL 60637
| | - Changgeng Ruan
- Jiangsu Institute of Hematology, The First Affiliated Hospital and Collaborative Innovation Center of Hematology, State Key Laboraotry of Radiation Medicine and Protection, Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Suzhou, Jiangsu 215006, China
| | - Kesheng Dai
- Jiangsu Institute of Hematology, The First Affiliated Hospital and Collaborative Innovation Center of Hematology, State Key Laboraotry of Radiation Medicine and Protection, Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Suzhou, Jiangsu 215006, China;
| |
Collapse
|
78
|
Hegde S, Akbar H, Zheng Y, Cancelas JA. Towards increasing shelf life and haemostatic potency of stored platelet concentrates. Curr Opin Hematol 2018; 25:500-508. [PMID: 30281037 PMCID: PMC6532779 DOI: 10.1097/moh.0000000000000456] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Platelet transfusion is a widely used therapy in treating or preventing bleeding and haemorrhage in patients with thrombocytopenia or trauma. Compared with the relative ease of platelet transfusion, current practice for the storage of platelets is inefficient, costly and relatively unsafe, with platelets stored at room temperature (RT) for upto 5-7 days. RECENT FINDINGS During storage, especially at cold temperatures, platelets undergo progressive and deleterious changes, collectively termed the 'platelet storage lesion', which decrease their haemostatic function and posttransfusion survival. Recent progress in understanding platelet activation and host clearance mechanisms is leading to the consideration of both old and novel storage conditions that use refrigeration and/or cryopreservation to overcome various storage lesions and significantly extend platelet shelf-life with a reduced risk of pathogen contamination. SUMMARY A review of the advantages and disadvantages of alternative methods for platelet storage is presented from both a clinical and biological perspective. It is anticipated that future platelet preservation involving cold, frozen and/or pathogen reduction strategies in a proper platelet additive solution will enable longer term and safer platelet storage.
Collapse
Affiliation(s)
- Shailaja Hegde
- Hoxworth Blood Center, University of Cincinnati Academic Health Center
| | - Huzoor Akbar
- Division of Experimental Hematology and Cancer Biology, Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA
| | - Yi Zheng
- Division of Experimental Hematology and Cancer Biology, Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati
| | - Jose A. Cancelas
- Hoxworth Blood Center, University of Cincinnati Academic Health Center
- Division of Experimental Hematology and Cancer Biology, Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati
| |
Collapse
|
79
|
Abonnenc M, Tissot JD, Prudent M. General overview of blood products in vitro quality: Processing and storage lesions. Transfus Clin Biol 2018; 25:269-275. [PMID: 30241785 DOI: 10.1016/j.tracli.2018.08.162] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 08/29/2018] [Indexed: 12/12/2022]
Abstract
Blood products are issued from blood collection. Collected blood is immediately mixed with anticoagulant solutions that immediately induce chemical and/or biochemical modifications. Collected blood is then transformed into different blood products according to various steps of fabrication. All these steps induce either reversible or irreversible "preparation-related" lesions that combine with "storage-related" lesions. This short paper aims to provide an overview of the alterations that are induced by the "non-physiological" processes used to prepare blood products that are used in clinical practice.
Collapse
Affiliation(s)
- Mélanie Abonnenc
- Transfusion interrégionale CRS, laboratoire de recherche sur les produits sanguins, route de la Corniche 2, 1066 Epalinges, Switzerland
| | - Jean-Daniel Tissot
- Transfusion interrégionale CRS, laboratoire de recherche sur les produits sanguins, route de la Corniche 2, 1066 Epalinges, Switzerland; Faculté de biologie et de médecine, université de Lausanne, Lausanne, Switzerland
| | - Michel Prudent
- Transfusion interrégionale CRS, laboratoire de recherche sur les produits sanguins, route de la Corniche 2, 1066 Epalinges, Switzerland; Faculté de biologie et de médecine, université de Lausanne, Lausanne, Switzerland.
| |
Collapse
|
80
|
Affiliation(s)
- Joel S Bennett
- From the Hematology-Oncology Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia.
| |
Collapse
|
81
|
Humbrecht C, Kientz D, Gachet C. Platelet transfusion: Current challenges. Transfus Clin Biol 2018; 25:151-164. [PMID: 30037501 DOI: 10.1016/j.tracli.2018.06.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 06/21/2018] [Indexed: 12/29/2022]
Abstract
Since the late sixties, platelet concentrates are transfused to patients presenting with severe thrombocytopenia, platelet function defects, injuries, or undergoing surgery, to prevent the risk of bleeding or to treat actual hemorrhage. Current practices differ according to the country or even in different hospitals and teams. Although crucial advances have been made during the last decades, questions and debates still arise about the right doses to transfuse, the use of prophylactic or therapeutic strategies, the nature and quality of PC, the storage conditions, the monitoring of transfusion efficacy and the microbiological and immunological safety of platelet transfusion. Finally, new challenges are emerging with potential new platelet products, including cold stored or in vitro produced platelets. The most debated of these points are reviewed.
Collapse
Affiliation(s)
- C Humbrecht
- Établissement français du sang grand est, 85-87, boulevard Lobau, 54064 Nancy cedex, France.
| | - D Kientz
- Établissement français du sang grand est, 85-87, boulevard Lobau, 54064 Nancy cedex, France
| | - C Gachet
- Établissement français du sang grand est, 85-87, boulevard Lobau, 54064 Nancy cedex, France.
| |
Collapse
|
82
|
Cho J, Kim H, Song J, Cheong JW, Shin JW, Yang WI, Kim HO. Platelet storage induces accelerated desialylation of platelets and increases hepatic thrombopoietin production. J Transl Med 2018; 16:199. [PMID: 30021591 PMCID: PMC6052694 DOI: 10.1186/s12967-018-1576-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 07/13/2018] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Stored platelets undergo deleterious changes, referred to as platelet storage lesions (PSLs), which accelerate the desialylation of platelets and result in their phagocytosis and clearance by hepatic macrophages. Recent studies have reported that Ashwell-Morell receptor binds to desialylated platelets, thereby inducing hepatic thrombopoietin (TPO) production in a mouse model. Therefore, this study aimed to demonstrate these relationships between PSL and hepatic TPO production in human study. METHODS Platelet concentrates (PCs) were obtained from 5 healthy volunteers and the remaining were discarded samples from the blood bank. PCs were divided into two halves, and stored either at 22 or 4 °C. Experiments were conducted using serial samples. Desialylation was assessed using flow cytometry, and structural changes were visualized using electron microscopy. Following co-culture of HepG2 cells (HB-8065, ATCC) with isolated platelets, hepatic TPO production was determined using real-time quantitative polymerase chain reaction and the supernatant TPO level was measured using a Luminex kit. RESULTS For 5 days of storage duration, platelet counts were not influenced by the storage conditions, but the degree of desialylation was proportional to the storage duration. Significant changes in the platelet surface and structure according to storage conditions were noted in electron microscopy. HepG2 cells incubated with aged platelets expressed more TPO mRNA, and supernatant TPO levels were proportional to the storage duration. Refrigeration also influenced on the results of this study, but they were not statistically significant. CONCLUSIONS This is the first study to demonstrate that, in vitro, aging and refrigeration affect the integrity of human platelets, resulting in induction of hepatic TPO mRNA and protein expression.
Collapse
Affiliation(s)
- Jooyoung Cho
- Department of Laboratory Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722 South Korea
- Department of Laboratory Medicine, Yonsei University Wonju College of Medicine, Wonju, South Korea
| | - Hyunkyung Kim
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Jaewoo Song
- Department of Laboratory Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722 South Korea
| | - June-Won Cheong
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Jeong Won Shin
- Department of Laboratory Medicine, Soonchunhyang University School of Medicine, Seoul, South Korea
| | - Woo Ick Yang
- Department of Pathology, Yonsei University College of Medicine, Seoul, South Korea
| | - Hyun Ok Kim
- Department of Laboratory Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722 South Korea
| |
Collapse
|
83
|
Intrinsic apoptosis circumvents the functional decline of circulating platelets but does not cause the storage lesion. Blood 2018; 132:197-209. [DOI: 10.1182/blood-2017-11-816355] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Accepted: 05/07/2018] [Indexed: 01/21/2023] Open
Abstract
Key Points
BAK/BAX depletion in murine platelets reveals that intrinsic apoptosis is not required for the development of the platelet storage lesion. Restriction of platelet life span by intrinsic apoptosis is pivotal to maintain a functional, hemostatically reactive platelet population.
Collapse
|
84
|
Wang S, Jiang T, Fan Y, Zhao S. A proteomic approach reveals the variation in human platelet protein composition after storage at different temperatures. Platelets 2018; 30:403-412. [PMID: 29595346 DOI: 10.1080/09537104.2018.1453060] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cryopreservation can slow down the metabolism and decrease the risk of bacterial contamination. But, chilled platelets (PLTs) show a reduced period in circulation due to the rapid clearance by hepatic cells or spleen macrophages after transfusion. The deleterious changes that PLTs undergo are mainly considered the result of PLT protein variation. However, the basis for proteomic variation of stored PLTs remains poorly understood. Besides count, activation markers (CD62P and Annexin V), and aggregation, we used quantitative mass spectrometry to create the first comprehensive and quantitative human PLT proteome of samples stored at different temperatures (22°C, 10°C and -80°C). We found different conditions caused different platelet storage lesion (PSL). PLT count was decreased no matter at what temperature stored. PLTs viability at low temperature dropped by 21.78% and 11.21%, respectively, as compared 10.26% at room temperature, there were no significant differences between the storage methods. Membrane expression of CD62P gradually increased in all groups especially stored at 22°C up to 40% and 10°C up to 30%. However, exposure of PS on the PLT membrane was below 1% in every group. The PLT proteome showed there were 575 and 454 potential proteins identified by general iTRAQ analysis and phosphorylation iTRAQ a nalysis, respectively, among them, 33 common differentially expressed proteins caused by storage time and 44 caused by storage temperature Especially, membrane-bound proteins (such as FERMT3, STX4, MYL9 and TAGLN2) played key roles in PLT storage lesion. The pathways "Endocytosis", "Fc gamma R-mediated phagocytosis" and "Regulation of actin cytoskeleton" were affected predominantly by storage time. And the pathways "SNARE interactions in vesicular transport" and "Vasopressin-regulated water reabsorption" were affected by cold storage in our study. Proteomic results can help us to understand PLT biochemistry and physiology and thus unravel the mechanisms of PSL in time and space for more successful PLT transfusion therapy.
Collapse
Affiliation(s)
- Shichun Wang
- a Department of Blood Transfusion , Southwest Hospital, the Third Military Medical University , Chongqing , P.R. China
| | - Tianlun Jiang
- a Department of Blood Transfusion , Southwest Hospital, the Third Military Medical University , Chongqing , P.R. China
| | - Yahan Fan
- a Department of Blood Transfusion , Southwest Hospital, the Third Military Medical University , Chongqing , P.R. China
| | - Shuming Zhao
- a Department of Blood Transfusion , Southwest Hospital, the Third Military Medical University , Chongqing , P.R. China
| |
Collapse
|
85
|
Mechanisms of platelet clearance and translation to improve platelet storage. Blood 2018; 131:1512-1521. [PMID: 29475962 DOI: 10.1182/blood-2017-08-743229] [Citation(s) in RCA: 150] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 01/28/2018] [Indexed: 02/01/2023] Open
Abstract
Hundreds of billions of platelets are cleared daily from circulation via efficient and highly regulated mechanisms. These mechanisms may be stimulated by exogenous reagents or environmental changes to accelerate platelet clearance, leading to thrombocytopenia. The interplay between antiapoptotic Bcl-xL and proapoptotic molecules Bax and Bak sets an internal clock for the platelet lifespan, and BH3-only proteins, mitochondrial permeabilization, and phosphatidylserine (PS) exposure may also contribute to apoptosis-induced platelet clearance. Binding of plasma von Willebrand factor or antibodies to the ligand-binding domain of glycoprotein Ibα (GPIbα) on platelets can activate GPIb-IX in a shear-dependent manner by inducing unfolding of the mechanosensory domain therein, and trigger downstream signaling in the platelet including desialylation and PS exposure. Deglycosylated platelets are recognized by the Ashwell-Morell receptor and potentially other scavenger receptors, and are rapidly cleared by hepatocytes and/or macrophages. Inhibitors of platelet clearance pathways, including inhibitors of GPIbα shedding, neuraminidases, and platelet signaling, are efficacious at preserving the viability of platelets during storage and improving their recovery and survival in vivo. Overall, common mechanisms of platelet clearance have begun to emerge, suggesting potential strategies to extend the shelf-life of platelets stored at room temperature or to enable refrigerated storage.
Collapse
|
86
|
Waters L, Cameron M, Padula MP, Marks DC, Johnson L. Refrigeration, cryopreservation and pathogen inactivation: an updated perspective on platelet storage conditions. Vox Sang 2018; 113:317-328. [PMID: 29441601 DOI: 10.1111/vox.12640] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 11/28/2017] [Accepted: 01/15/2018] [Indexed: 01/08/2023]
Abstract
Conventional storage of platelet concentrates limits their shelf life to between 5 and 7 days due to the risk of bacterial proliferation and the development of the platelet storage lesion. Cold storage and cryopreservation of platelets may facilitate extension of the shelf life to weeks and years, and may also provide the benefit of being more haemostatically effective than conventionally stored platelets. Further, treatment of platelet concentrates with pathogen inactivation systems reduces bacterial contamination and provides a safeguard against the risk of emerging and re-emerging pathogens. While each of these alternative storage techniques is gaining traction individually, little work has been done to examine the effect of combining treatments in an effort to further improve product safety and minimize wastage. This review aims to discuss the benefits of alternative storage techniques and how they may be combined to alleviate the problems associated with conventional platelet storage.
Collapse
Affiliation(s)
- L Waters
- Research & Development, Australian Red Cross Blood Service, Alexandria, NSW, Australia.,School of Life Sciences, University of Technology Sydney, Sydney, NSW, Australia
| | - M Cameron
- Research & Development, Australian Red Cross Blood Service, Alexandria, NSW, Australia.,School of Life Sciences, University of Technology Sydney, Sydney, NSW, Australia
| | - M P Padula
- School of Life Sciences, University of Technology Sydney, Sydney, NSW, Australia
| | - D C Marks
- Research & Development, Australian Red Cross Blood Service, Alexandria, NSW, Australia
| | - L Johnson
- Research & Development, Australian Red Cross Blood Service, Alexandria, NSW, Australia
| |
Collapse
|
87
|
Handtke S, Steil L, Greinacher A, Thiele T. Toward the Relevance of Platelet Subpopulations for Transfusion Medicine. Front Med (Lausanne) 2018; 5:17. [PMID: 29459897 PMCID: PMC5807390 DOI: 10.3389/fmed.2018.00017] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 01/18/2018] [Indexed: 12/11/2022] Open
Abstract
Circulating platelets consist of subpopulations with different age, maturation state and size. In this review, we address the association between platelet size and platelet function and summarize the current knowledge on platelet subpopulations including reticulated platelets, procoagulant platelets and platelets exposing signals to mediate their clearance. Thereby, we emphasize the impact of platelet turnover as an important condition for platelet production in vivo. Understanding of the features that characterize platelet subpopulations is very relevant for the methods of platelet concentrate production, which may enrich or deplete particular platelet subpopulations. Moreover, the concept of platelet size being associated with platelet function may be attractive for transfusion medicine as it holds the perspective to separate platelet subpopulations with specific functional capabilities.
Collapse
Affiliation(s)
- Stefan Handtke
- Institut für Immunologie und Transfusionsmedizin, Greifswald, Germany
| | - Leif Steil
- Interfakultäres Institut für Funktionelle Genomforschung, Greifswald, Germany
| | | | - Thomas Thiele
- Institut für Immunologie und Transfusionsmedizin, Greifswald, Germany
| |
Collapse
|
88
|
Yang J, Yin W, Zhang Y, Sun Y, Ma T, Gu S, Gao Y, Zhang X, Yuan J, Wang W. Evaluation of the advantages of platelet concentrates stored at 4°C versus 22°C. Transfusion 2017; 58:736-747. [PMID: 29266276 DOI: 10.1111/trf.14462] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 10/30/2017] [Accepted: 11/12/2017] [Indexed: 01/23/2023]
Affiliation(s)
- Jiangcun Yang
- Department of Transfusion Medicine, Shaanxi Provincial People's Hospital; Xi'an China
| | - Wen Yin
- Department of Transfusion Medicine; Xijing Hospital, the Fourth Military Medical University; Xi'an China
| | - Yali Zhang
- Department of Blood Components Preparation; Shaanxi Provincial Blood Center; Xi'an China
| | - Yang Sun
- Department of Transfusion Medicine, Shaanxi Provincial People's Hospital; Xi'an China
| | - Ting Ma
- Department of Transfusion Medicine, Shaanxi Provincial People's Hospital; Xi'an China
| | - Shunli Gu
- Department of Transfusion Medicine; Xijing Hospital, the Fourth Military Medical University; Xi'an China
| | - Ying Gao
- Department of Hematology, Shaanxi Provincial People's Hospital; Xi'an China
| | - Xiaole Zhang
- Department of Transfusion Medicine; Xijing Hospital, the Fourth Military Medical University; Xi'an China
| | - Jun Yuan
- Department of Laboratory; Shaanxi Provincial People's Hospital; Xi'an China
| | - Wenhua Wang
- Department of Transfusion Medicine, Shaanxi Provincial People's Hospital; Xi'an China
| |
Collapse
|
89
|
Chen W, Druzak SA, Wang Y, Josephson CD, Hoffmeister KM, Ware J, Li R. Refrigeration-Induced Binding of von Willebrand Factor Facilitates Fast Clearance of Refrigerated Platelets. Arterioscler Thromb Vasc Biol 2017; 37:2271-2279. [PMID: 29097365 DOI: 10.1161/atvbaha.117.310062] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 10/23/2017] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Apheresis platelets for transfusion treatment are currently stored at room temperature because after refrigeration platelets are rapidly cleared on transfusion. In this study, the role of von Willebrand factor (VWF) in the clearance of refrigerated platelets is addressed. APPROACH AND RESULTS Human and murine platelets were refrigerated in gas-permeable bags at 4°C for 24 hours. VWF binding, platelet signaling events, and platelet post-transfusion recovery and survival were measured. After refrigeration, the binding of plasma VWF to platelets was drastically increased, confirming earlier studies. The binding was blocked by peptide OS1 that bound specifically to platelet glycoprotein (GP)Ibα and was absent in VWF-/- plasma. Although surface expression of GPIbα was reduced after refrigeration, refrigeration-induced VWF binding under physiological shear induced unfolding of the GPIbα mechanosensory domain on the platelet, as evidenced by increased exposure of a linear epitope therein. Refrigeration and shear treatment also induced small elevation of intracellular Ca2+, phosphatidylserine exposure, and desialylation of platelets, which were absent in VWF-/- platelets or inhibited by OS1, which is a monomeric 11-residue peptide (CTERMALHNLC). Furthermore, refrigerated VWF-/- platelets displayed increased post-transfusion recovery and survival than wild-type ones. Similarly, adding OS1 to transgenic murine platelets expressing only human GPIbα during refrigeration improved their post-transfusion recovery and survival. CONCLUSIONS Refrigeration-induced binding of VWF to platelets facilitates their rapid clearance by inducing GPIbα-mediated signaling. Our results suggest that inhibition of the VWF-GPIbα interaction may be a potential strategy to enable refrigeration of platelets for transfusion treatment.
Collapse
Affiliation(s)
- Wenchun Chen
- From the Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, GA (W.C., S.A.D., Y.W., C.D.J., R.L.); Department of Pediatrics (W.C., S.A.D., Y.W., C.D.J., R.L.) and Department of Pathology (C.D.J.), Emory University School of Medicine, Atlanta, GA; Blood Research Institute, BloodCenter of Wisconsin, Milwaukee (K.M.H.); and Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock (J.W.)
| | - Samuel A Druzak
- From the Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, GA (W.C., S.A.D., Y.W., C.D.J., R.L.); Department of Pediatrics (W.C., S.A.D., Y.W., C.D.J., R.L.) and Department of Pathology (C.D.J.), Emory University School of Medicine, Atlanta, GA; Blood Research Institute, BloodCenter of Wisconsin, Milwaukee (K.M.H.); and Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock (J.W.)
| | - Yingchun Wang
- From the Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, GA (W.C., S.A.D., Y.W., C.D.J., R.L.); Department of Pediatrics (W.C., S.A.D., Y.W., C.D.J., R.L.) and Department of Pathology (C.D.J.), Emory University School of Medicine, Atlanta, GA; Blood Research Institute, BloodCenter of Wisconsin, Milwaukee (K.M.H.); and Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock (J.W.)
| | - Cassandra D Josephson
- From the Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, GA (W.C., S.A.D., Y.W., C.D.J., R.L.); Department of Pediatrics (W.C., S.A.D., Y.W., C.D.J., R.L.) and Department of Pathology (C.D.J.), Emory University School of Medicine, Atlanta, GA; Blood Research Institute, BloodCenter of Wisconsin, Milwaukee (K.M.H.); and Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock (J.W.)
| | - Karin M Hoffmeister
- From the Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, GA (W.C., S.A.D., Y.W., C.D.J., R.L.); Department of Pediatrics (W.C., S.A.D., Y.W., C.D.J., R.L.) and Department of Pathology (C.D.J.), Emory University School of Medicine, Atlanta, GA; Blood Research Institute, BloodCenter of Wisconsin, Milwaukee (K.M.H.); and Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock (J.W.)
| | - Jerry Ware
- From the Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, GA (W.C., S.A.D., Y.W., C.D.J., R.L.); Department of Pediatrics (W.C., S.A.D., Y.W., C.D.J., R.L.) and Department of Pathology (C.D.J.), Emory University School of Medicine, Atlanta, GA; Blood Research Institute, BloodCenter of Wisconsin, Milwaukee (K.M.H.); and Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock (J.W.)
| | - Renhao Li
- From the Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, GA (W.C., S.A.D., Y.W., C.D.J., R.L.); Department of Pediatrics (W.C., S.A.D., Y.W., C.D.J., R.L.) and Department of Pathology (C.D.J.), Emory University School of Medicine, Atlanta, GA; Blood Research Institute, BloodCenter of Wisconsin, Milwaukee (K.M.H.); and Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock (J.W.).
| |
Collapse
|
90
|
Jones AR, Frazier SK. Consequences of Transfusing Blood Components in Patients With Trauma: A Conceptual Model. Crit Care Nurse 2017; 37:18-30. [PMID: 28365647 DOI: 10.4037/ccn2017965] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
Transfusion of blood components is often required in resuscitation of patients with major trauma. Packed red blood cells and platelets break down and undergo chemical changes during storage (known as the storage lesion) that lead to an inflammatory response once the blood components are transfused to patients. Although some evidence supports a detrimental association between transfusion and a patient's outcome, the mechanisms connecting transfusion of stored components to outcomes remain unclear. The purpose of this review is to provide critical care nurses with a conceptual model to facilitate understanding of the relationship between the storage lesion and patients' outcomes after trauma; outcomes related to trauma, hemorrhage, and blood component transfusion are grouped according to those occurring in the short-term (≤30 days) and the long-term (>30 days). Complete understanding of these clinical implications is critical for practitioners in evaluating and treating patients given transfusions after traumatic injury.
Collapse
Affiliation(s)
- Allison R Jones
- Allison R. Jones is an assistant professor, Department of Acute, Chronic, and Continuing Care, School of Nursing, University of Alabama, Birmingham, Alabama. She has a clinical background in emergency and trauma nursing. In research, she focuses on the consequences of blood component storage and transfusion, with particular interest in transfusion after trauma. .,Susan K. Frazier is the director of the PhD program, a codirector of the RICH Heart Program, and an associate professor, College of Nursing, University of Kentucky, Lexington, Kentucky. Her research focuses on cardiopulmonary interactions in a variety of critically ill patients, including patients with acute heart failure, acute decompensated heart failure, acute respiratory distress syndrome, chronic obstructive pulmonary disease, and multiple trauma.
| | - Susan K Frazier
- Allison R. Jones is an assistant professor, Department of Acute, Chronic, and Continuing Care, School of Nursing, University of Alabama, Birmingham, Alabama. She has a clinical background in emergency and trauma nursing. In research, she focuses on the consequences of blood component storage and transfusion, with particular interest in transfusion after trauma.,Susan K. Frazier is the director of the PhD program, a codirector of the RICH Heart Program, and an associate professor, College of Nursing, University of Kentucky, Lexington, Kentucky. Her research focuses on cardiopulmonary interactions in a variety of critically ill patients, including patients with acute heart failure, acute decompensated heart failure, acute respiratory distress syndrome, chronic obstructive pulmonary disease, and multiple trauma
| |
Collapse
|
91
|
NasrEldin E. Effect of cold storage on platelets quality stored in a small containers: Implications for pediatric transfusion. PEDIATRIC HEMATOLOGY ONCOLOGY JOURNAL 2017. [DOI: 10.1016/j.phoj.2017.07.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
92
|
Twinfilin 2a regulates platelet reactivity and turnover in mice. Blood 2017; 130:1746-1756. [PMID: 28743718 DOI: 10.1182/blood-2017-02-770768] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 07/21/2017] [Indexed: 01/22/2023] Open
Abstract
Regulated reorganization of the actin cytoskeleton is a prerequisite for proper platelet production and function. Consequently, defects in proteins controlling actin dynamics have been associated with platelet disorders in humans and mice. Twinfilin 2a (Twf2a) is a small actin-binding protein that inhibits actin filament assembly by sequestering actin monomers and capping filament barbed ends. Moreover, Twf2a binds heterodimeric capping proteins, but the role of this interaction in cytoskeletal dynamics has remained elusive. Even though Twf2a has pronounced effects on actin dynamics in vitro, only little is known about its function in vivo. Here, we report that constitutive Twf2a-deficient mice (Twf2a-/-) display mild macrothrombocytopenia due to a markedly accelerated platelet clearance in the spleen. Twf2a-/- platelets showed enhanced integrin activation and α-granule release in response to stimulation of (hem) immunoreceptor tyrosine-based activation motif (ITAM) and G-protein-coupled receptors, increased adhesion and aggregate formation on collagen I under flow, and accelerated clot retraction and spreading on fibrinogen. In vivo, Twf2a deficiency resulted in shortened tail bleeding times and faster occlusive arterial thrombus formation. The hyperreactivity of Twf2a-/- platelets was attributed to enhanced actin dynamics, characterized by an increased activity of n-cofilin and profilin 1, leading to a thickened cortical cytoskeleton and hence sustained integrin activation by limiting calpain-mediated integrin inactivation. In summary, our results reveal the first in vivo functions of mammalian Twf2a and demonstrate that Twf2a-controlled actin rearrangements dampen platelet activation responses in a n-cofilin- and profilin 1-dependent manner, thereby indirectly regulating platelet reactivity and half-life in mice.
Collapse
|
93
|
Stivala S, Gobbato S, Infanti L, Reiner MF, Bonetti N, Meyer SC, Camici GG, Lüscher TF, Buser A, Beer JH. Amotosalen/ultraviolet A pathogen inactivation technology reduces platelet activatability, induces apoptosis and accelerates clearance. Haematologica 2017; 102:1650-1660. [PMID: 28729303 PMCID: PMC5622849 DOI: 10.3324/haematol.2017.164137] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 07/13/2017] [Indexed: 01/03/2023] Open
Abstract
Amotosalen and ultraviolet A (UVA) photochemical-based pathogen reduction using the Intercept™ Blood System (IBS) is an effective and established technology for platelet and plasma components, which is adopted in more than 40 countries worldwide. Several reports point towards a reduced platelet function after Amotosalen/UVA exposure. The study herein was undertaken to identify the mechanisms responsible for the early impairment of platelet function by the IBS. Twenty-five platelet apheresis units were collected from healthy volunteers following standard procedures and split into 2 components, 1 untreated and the other treated with Amotosalen/UVA. Platelet impedance aggregation in response to collagen and thrombin was reduced by 80% and 60%, respectively, in IBS-treated units at day 1 of storage. Glycoprotein Ib (GpIb) levels were significantly lower in IBS samples and soluble glycocalicin correspondingly augmented; furthermore, GpIbα was significantly more desialylated as shown by Erythrina Cristagalli Lectin (ECL) binding. The pro-apoptotic Bak protein was significantly increased, as well as the MAPK p38 phosphorylation and caspase-3 cleavage. Stored IBS-treated platelets injected into immune-deficient nonobese diabetic/severe combined immunodeficiency (NOD/SCID) mice showed a faster clearance. We conclude that the IBS induces platelet p38 activation, GpIb shedding and platelet apoptosis through a caspase-dependent mechanism, thus reducing platelet function and survival. These mechanisms are of relevance in transfusion medicine, where the IBS increases patient safety at the expense of platelet function and survival.
Collapse
Affiliation(s)
- Simona Stivala
- Laboratory for Platelet Research, Center for Molecular Cardiology, University of Zurich, Switzerland.,Department of Internal Medicine, Cantonal Hospital Baden, Switzerland
| | - Sara Gobbato
- Laboratory for Platelet Research, Center for Molecular Cardiology, University of Zurich, Switzerland.,Department of Internal Medicine, Cantonal Hospital Baden, Switzerland
| | - Laura Infanti
- Regional Blood Transfusion Service of the Swiss Red Cross, Basel, Switzerland
| | - Martin F Reiner
- Laboratory for Platelet Research, Center for Molecular Cardiology, University of Zurich, Switzerland.,Department of Internal Medicine, Cantonal Hospital Baden, Switzerland
| | - Nicole Bonetti
- Laboratory for Platelet Research, Center for Molecular Cardiology, University of Zurich, Switzerland.,Department of Internal Medicine, Cantonal Hospital Baden, Switzerland
| | - Sara C Meyer
- Division of Hematology and Department of Biomedicine, University Hospital Basel, Switzerland
| | | | - Thomas F Lüscher
- Department of Cardiology, University Heart Center, University Hospital Zurich, Switzerland
| | - Andreas Buser
- Regional Blood Transfusion Service of the Swiss Red Cross, Basel, Switzerland
| | - Jürg H Beer
- Laboratory for Platelet Research, Center for Molecular Cardiology, University of Zurich, Switzerland .,Department of Internal Medicine, Cantonal Hospital Baden, Switzerland
| |
Collapse
|
94
|
Sialylation on O-glycans protects platelets from clearance by liver Kupffer cells. Proc Natl Acad Sci U S A 2017; 114:8360-8365. [PMID: 28716912 DOI: 10.1073/pnas.1707662114] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Most platelet membrane proteins are modified by mucin-type core 1-derived glycans (O-glycans). However, the biological importance of O-glycans in platelet clearance is unclear. Here, we generated mice with a hematopoietic cell-specific loss of O-glycans (HC C1galt1-/- ). These mice lack O-glycans on platelets and exhibit reduced peripheral platelet numbers. Platelets from HC C1galt1-/- mice show reduced levels of α-2,3-linked sialic acids and increased accumulation in the liver relative to wild-type platelets. The preferential accumulation of HC C1galt1-/- platelets in the liver was reduced in mice lacking the hepatic asialoglycoprotein receptor [Ashwell-Morell receptor (AMR)]. However, we found that Kupffer cells are the primary cells phagocytosing HC C1galt1-/- platelets in the liver. Our results demonstrate that hepatic AMR promotes preferential adherence to and phagocytosis of desialylated and/or HC C1galt1-/- platelets by the Kupffer cell through its C-type lectin receptor CLEC4F. These findings provide insights into an essential role for core 1 O-glycosylation of platelets in their clearance in the liver.
Collapse
|
95
|
Abstract
PURPOSE OF REVIEW This review seeks to describe the role of the asialoglycoprotein receptor (ASGR) in modulating non-HDL lipoprotein levels, platelet numbers and atherosclerosis. RECENT FINDINGS Genetics studies have revealed that ASGR haplodeficiency provides protection from atherosclerosis. The potential interactions of ASGR with LDL receptor may regulate the rate of LDL uptake and as a result may lower plasma non-HDL cholesterol. ASGR clears senescent platelets and induces the expression of hepatic thrombopoietin. Platelet activity promotes plaque formation and atherosclerosis. SUMMARY ASGR is a multifunctional receptor specializing in clearance of desialylated glycoproteins and platelets. The impact of ASGR on the levels of plasma non-HDL cholesterol makes it a potential therapeutic target for the treatment of hypercholesterolemia and atherosclerosis.
Collapse
Affiliation(s)
- Suleiman A Igdoura
- Departments of Biology and Pathology, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
96
|
Abstract
A wide variety of clinical conditions, associated with low circulating platelet counts, require platelet transfusion in order to normalize hemostatic function. Although single-donor apheresis platelets bear the lowest risk of transfusion-transmitted infections, pathogen reduction technologies (PRT) are being implemented worldwide to reduce this risk further through inactivation of known, emergent and as yet to be discovered nucleic acid-based pathogens. Human blood platelets are now known to harbor a diverse transcriptome, important to their function and comprised of >5000 protein-coding messenger RNAs and different classes of non-coding RNAs, including microRNAs. Our appreciation of the nucleic acid-dependent functions of platelets is likely to increase. On the other hand, the side effects of PRT on platelet function are underappreciated. Recent evidences suggest that PRT may compromise platelets' responsiveness to agonists, and induce platelet activation. For instance, platelets have the propensity to release proinflammatory microparticles (MPs) upon activation, and the possibility that PRT may enhance the production of platelet MPs in platelet concentrates (PCs) appears likely. With this in mind, it would be timely and appropriate to investigate other means to inactivate pathogens more specifically, or to modify the currently available PRT so to better preserve the platelet function and improve the safety of PCs; platelets' perspective to PRT deserves to be considered.
Collapse
Affiliation(s)
- Abdimajid Osman
- a Department of Clinical Chemistry , Region Östergötland , Linköping , Sweden.,b Department of Clinical and Experimental Medicine , University of Linköping , Linköping , Sweden
| | - Walter E Hitzler
- c Transfusion Center, University Medical Center of the Johannes Gutenberg University Mainz , Hochhaus Augustusplatz , Mainz , Germany
| | - Patrick Provost
- d CHUQ Research Center/CHUL , 2705 Blvd Laurier, Quebec , QC , Canada.,e Faculty of Medicine , Université Laval , Quebec , QC , Canada
| |
Collapse
|
97
|
Platelet clearance by the hepatic Ashwell-Morrell receptor: mechanisms and biological significance. Thromb Res 2017; 141 Suppl 2:S68-72. [PMID: 27207430 DOI: 10.1016/s0049-3848(16)30370-x] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The daily production of billions of platelets must be regulated to avoid spontaneous bleeding or arterial occlusion and organ damage. Complex mechanisms control platelet production and clearance in physiological and pathological conditions. This review will focus on the mechanisms of platelet senescence with specific emphasis on the role of post-translational modifications in platelet life-span and thrombopoietin production downstream of the hepatic Ashwell-Morrell receptor.
Collapse
|
98
|
Temperature effects on the activity, shape, and storage of platelets from 13-lined ground squirrels. J Comp Physiol B 2017; 187:815-825. [PMID: 28332020 DOI: 10.1007/s00360-017-1081-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 12/15/2016] [Accepted: 02/26/2017] [Indexed: 01/14/2023]
Abstract
The objective of this study is to determine how a hibernating mammal avoids the formation of blood clots under periods of low blood flow. A microfluidic vascular injury model was performed to differentiate the effects of temperature and shear rate on platelet adhesion to collagen. Human and ground squirrel whole blood was incubated at 15 or 37 °C and then passed through a microfluidic chamber over a 250-µm strip of type I fibrillar collagen at that temperature and the shear rates of 50 or 300 s-1 to simulate torpid and aroused conditions, respectively. At 15 °C, both human and ground squirrel platelets showed a 90-95% decrease in accumulation on collagen independent of shear rate. At 37 °C, human platelet accumulation reduced by 50% at 50 s-1 compared to 300 s-1, while ground squirrel platelet accumulation dropped by 80%. When compared to platelets from non-hibernating animals, platelets from animals collected after arousal from torpor showed a 60% decrease in binding at 37 °C and 300 s-1, but a 2.5-fold increase in binding at 15 °C and 50 s-1. vWF binding in platelets from hibernating ground squirrels was decreased by 50% relative to non-hibernating platelets. The source of the plasma that platelets were stored in did not affect the results indicating that the decreased vWF binding was a property of the platelets. Upon chilling, ground squirrel platelets increase microtubule assembly leading to the formation of long rods. This shape change is concurrent with sequestration of platelets in the liver and not the spleen. In conclusion, it appears that ground squirrel platelets are sequestered in the liver during torpor and have reduced binding capacity for plasma vWF and lower accumulation on collagen at low shear rates and after storage at cold temperatures, while still being activated by external agonists. These adaptations would protect the animals from spontaneous thrombus formation during torpor but allow them to restore normal platelet function upon arousal.
Collapse
|
99
|
Braune S, Fröhlich GM, Lendlein A, Jung F. Effect of temperature on platelet adherence. Clin Hemorheol Microcirc 2017; 61:681-8. [PMID: 26639771 DOI: 10.3233/ch-152028] [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: 11/15/2022]
Abstract
BACKGROUND Thrombogenicity is one of the main parameters tested in vitro to evaluate the hemocompatibility of artificial surfaces. While the influence of the temperature on platelet aggregation has been addressed by several studies, the temperature influence on the adherence of platelets to body foreign surfaces as an important aspect of biomedical device handling has not yet been explored. Therefore, we analyzed the influence of two typically applied incubation-temperatures (22°C and 37°C) on the adhesion of platelets to biomaterials. MATERIAL AND METHODS Thrombogenicity of three different polymers - medical grade poly(dimethyl siloxane) (PDMS), polytetrafluoroethylene (PTFE) and polyethylene terephthalate (PET) - were studied in an in vitro static test. Platelet adhesion was studied with stringently characterized blood from apparently healthy subjects. Collection of whole blood and preparation of platelet rich plasma (PRP) was carried out at room temperature (22°C). PRP was incubated with the polymers either at 22°C or 37°C. Surface adherent platelets were fixed, fluorescently labelled and assessed by an image-based approach. RESULTS AND DISCUSSION Differences in the density of adherent platelets after incubation at 22°C and 37°C occurred on PDMS and PET. Similar levels of adherent platelets were observed on the very thrombogenic PTFE. The covered surface areas per single platelet were analyzed to measure the state of platelet activation and revealed no differences between the two incubation temperatures for any of the analyzed polymers. Irrespective of the observed differences between the low and medium thrombogenic PDMS and PET and the higher variability at 22°C, the thrombogenicity of the three investigated polymers was evaluated being comparable at both incubation temperatures.
Collapse
Affiliation(s)
- S Braune
- Institute of Biomaterial Science, Helmholtz-Zentrum Geesthacht, Teltow, Germany.,Berlin-Brandenburg Center for Regenerative Therapies, Teltow and Berlin, Germany
| | | | - A Lendlein
- Institute of Biomaterial Science, Helmholtz-Zentrum Geesthacht, Teltow, Germany.,Berlin-Brandenburg Center for Regenerative Therapies, Teltow and Berlin, Germany.,Institute of Chemistry, University of Potsdam, Potsdam, Germany
| | - F Jung
- Institute of Biomaterial Science, Helmholtz-Zentrum Geesthacht, Teltow, Germany.,Berlin-Brandenburg Center for Regenerative Therapies, Teltow and Berlin, Germany
| |
Collapse
|
100
|
Acker JP, Marks DC, Sheffield WP. Quality Assessment of Established and Emerging Blood Components for Transfusion. JOURNAL OF BLOOD TRANSFUSION 2016; 2016:4860284. [PMID: 28070448 PMCID: PMC5192317 DOI: 10.1155/2016/4860284] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 11/02/2016] [Indexed: 12/16/2022]
Abstract
Blood is donated either as whole blood, with subsequent component processing, or through the use of apheresis devices that extract one or more components and return the rest of the donation to the donor. Blood component therapy supplanted whole blood transfusion in industrialized countries in the middle of the twentieth century and remains the standard of care for the majority of patients receiving a transfusion. Traditionally, blood has been processed into three main blood products: red blood cell concentrates; platelet concentrates; and transfusable plasma. Ensuring that these products are of high quality and that they deliver their intended benefits to patients throughout their shelf-life is a complex task. Further complexity has been added with the development of products stored under nonstandard conditions or subjected to additional manufacturing steps (e.g., cryopreserved platelets, irradiated red cells, and lyophilized plasma). Here we review established and emerging methodologies for assessing blood product quality and address controversies and uncertainties in this thriving and active field of investigation.
Collapse
Affiliation(s)
- Jason P. Acker
- Centre for Innovation, Canadian Blood Services, Edmonton, AB, Canada
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - Denese C. Marks
- Research and Development, Australian Red Cross Blood Service, Sydney, NSW, Australia
| | - William P. Sheffield
- Centre for Innovation, Canadian Blood Services, Hamilton, ON, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|