51
|
Tohidi-Esfahani I, Lee CSM, Liang HPH, Chen VMY. Procoagulant platelets: Laboratory detection and clinical significance. Int J Lab Hematol 2021; 42 Suppl 1:59-67. [PMID: 32543068 DOI: 10.1111/ijlh.13197] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 03/10/2020] [Indexed: 12/15/2022]
Abstract
Platelets play a critical role in both haemostasis and thrombosis, and it is now evident that not all platelets behave the same when they are called to action. A functionally distinct subpopulation of platelets forms in response to maximal agonist stimulation: the procoagulant platelet. This platelet subpopulation is defined by its ability to expose phosphatidylserine on its surface, allowing for coagulation factor complexes to form and generate bursts of thrombin and fibrin to stabilize platelet clots. Reduced levels of procoagulant platelets have been linked to bleeding in Scott's syndrome and haemophilia A patients, and elevated levels have been demonstrated in many thrombotic disorders, including identifying patients at higher risk for stroke recurrence. One obstacle for incorporating an assay for measuring procoagulant platelets into clinical management algorithms is the lack of consensus on the exact definition and markers for this subpopulation. This review will outline the biological characteristics of procoagulant platelets and the laboratory assays currently used to identify them in research settings. It will summarize the findings of clinical research demonstrating the relevance of measuring the procoagulant platelet levels in patients and will discuss how an appropriate assay can be used to elucidate the mechanism behind the formation of this subpopulation, facilitating novel drug discovery to improve upon current outcomes in cardiovascular and other thrombotic disorders.
Collapse
Affiliation(s)
- Ibrahim Tohidi-Esfahani
- ANZAC Research Institute, University of Sydney, Sydney, New South Wales, Australia.,Department of Haematology, Concord Repatriation General Hospital, Sydney, New South Wales, Australia
| | - Christine S M Lee
- ANZAC Research Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Hai Po H Liang
- ANZAC Research Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Vivien M Y Chen
- ANZAC Research Institute, University of Sydney, Sydney, New South Wales, Australia.,Department of Haematology, Concord Repatriation General Hospital, Sydney, New South Wales, Australia
| |
Collapse
|
52
|
Fernández DI, Kuijpers MJE, Heemskerk JWM. Platelet calcium signaling by G-protein coupled and ITAM-linked receptors regulating anoctamin-6 and procoagulant activity. Platelets 2020; 32:863-871. [PMID: 33356720 DOI: 10.1080/09537104.2020.1859103] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Most agonists stimulate platelet Ca2+ rises via G-protein coupled receptors (GPCRs) or ITAM-linked receptors (ILRs). Well studied are the GPCRs stimulated by the soluble agonists thrombin (PAR1, PAR4), ADP (P2Y1, P2Y12), and thromboxane A2 (TP), signaling via phospholipase (PLC)β isoforms. The platelet ILRs glycoprotein VI (GPVI), C-type lectin-like receptor 2 (CLEC2), and FcγRIIa are stimulated by adhesive ligands or antibody complexes and signal via tyrosine protein kinases and PLCγ isoforms. Marked differences exist between the GPCR- and ILR-induced Ca2+ signaling in: (i) dependency of tyrosine phosphorylation; (ii) oscillatory versus continued Ca2+ rises by mobilization from the endoplasmic reticulum; and (iii) smaller or larger role of extracellular Ca2+ entry via STIM1/ORAI1. Co-stimulation of both types of receptors, especially by thrombin (PAR1/4) and collagen (GPVI), leads to a highly enforced Ca2+ rise, involving mitochondrial Ca2+ release, which activates the ion and phospholipid channel, anoctamin-6. This highly Ca2+-dependent process causes swelling, ballooning, and phosphatidylserine expression, establishing a unique platelet population swinging between vital and necrotic (procoagulant 'zombie' platelets). Additionally, the high Ca2+ status of procoagulant platelets induces a set of additional events: (i) Ca2+ dependent cleavage of signaling proteins and receptors via calpain and ADAM isoforms; (ii) microvesiculation; (iii) enhanced coagulation factor binding; and (iv) fibrin-coat formation involving transglutaminases. Given the additive roles of GPCR and ILR in Ca2+ signal generation, high-throughput screening of biomolecules or small molecules based on Ca2+ flux measurements provides a promising way to find new inhibitors interfering with prolonged high Ca2+, phosphatidylserine expression, and hence platelet procoagulant activity.
Collapse
Affiliation(s)
- Delia I Fernández
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Marijke J E Kuijpers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Johan W M Heemskerk
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
53
|
Reddoch-Cardenas KM, Peltier GC, Chance TC, Nair PM, Meledeo MA, Ramasubramanian AK, Cap AP, Bynum JA. Cold storage of platelets in platelet additive solution maintains mitochondrial integrity by limiting initiation of apoptosis-mediated pathways. Transfusion 2020; 61:178-190. [PMID: 33294977 DOI: 10.1111/trf.16185] [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: 12/06/2019] [Revised: 07/03/2020] [Accepted: 07/08/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND Cold storage of platelets in plasma maintains hemostatic function and is an attractive alternative to room temperature platelets (RTPs). We have recently shown that functional differences between cold-stored platelets (CSPs) and RTPs after 5-day storage are associated with mitochondrial respiration and that CSPs in platelet (PLT) additive solution (PAS) can maintain hemostatic function for at least 15 days. STUDY DESIGN AND METHODS This study tested the hypothesis that cold storage in PAS preserves mitochondrial integrity by reducing PLT apoptosis. CSPs and RTPs in plasma or PAS were stored and assayed for up to 15 days for mitochondrial function and integrity, mitochondrial-associated mRNA transcript expression, apoptotic proteins, and apoptotic flow cytometry metrics. RESULTS CSP preserved mitochondria-associated mRNA comparable to baseline levels, improved mitochondrial respiration, and minimized depolarization to Day 15. Additionally, CSPs had minimal induction of caspases, preservation of plasma membrane integrity, and low expression of pro-apoptotic Bax. Storage in PAS appeared to be protective for RTPs in some parameters and enhanced the effects of CSPs. CONCLUSION Mitochondrial function and molecular analyses defined CSP priming as distinctly different from the well-documented RTP storage lesion. While current blood bank storage at room temperature is limited to 5 to 7 days, refrigeration and storage in PAS for up to 15 days may represent an opportunity to enhance inventories and access to PLT hemostatic support for bleeding patients.
Collapse
Affiliation(s)
| | - Grant C Peltier
- US Army Institute of Surgical Research, JBSA-Fort Sam Houston, Sam Houston, Texas, USA
| | | | - Prajeeda M Nair
- US Army Institute of Surgical Research, JBSA-Fort Sam Houston, Sam Houston, Texas, USA
| | - Michael A Meledeo
- US Army Institute of Surgical Research, JBSA-Fort Sam Houston, Sam Houston, Texas, USA
| | | | - Andrew P Cap
- US Army Institute of Surgical Research, JBSA-Fort Sam Houston, Sam Houston, Texas, USA
| | - James A Bynum
- US Army Institute of Surgical Research, JBSA-Fort Sam Houston, Sam Houston, Texas, USA
| |
Collapse
|
54
|
Sheriff J, Malone LE, Avila C, Zigomalas A, Bluestein D, Bahou WF. Shear-Induced Platelet Activation is Sensitive to Age and Calcium Availability: A Comparison of Adult and Cord Blood. Cell Mol Bioeng 2020; 13:575-590. [PMID: 33281988 PMCID: PMC7704822 DOI: 10.1007/s12195-020-00628-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 06/16/2020] [Indexed: 10/24/2022] Open
Abstract
INTRODUCTION Antiplatelet therapy for neonates and infants is often extrapolated from the adult experience, based on limited observation of agonist-induced neonatal platelet hypoactivity and poor understanding of flow shear-mediated platelet activation. Therefore, thrombotic events due to device-associated disturbed flow are inadequately mitigated in critically ill neonates with indwelling umbilical catheters and infants receiving cardiovascular implants. METHODS Whole blood (WB), platelet-rich plasma (PRP), and gel-filtered platelets (GFP) were prepared from umbilical cord and adult blood, and exposed to biochemical agonists or pathological shear stress of 70 dyne/cm2. We evaluated α-granule release, phosphatidylserine (PS) scrambling, and procoagulant response using P-selectin expression, Annexin V binding, and thrombin generation (PAS), respectively. Activation modulation due to depletion of intracellular and extracellular calcium, requisite second messengers, was also examined. RESULTS Similar P-selectin expression was observed for sheared adult and cord platelets, with concordant inhibition due to intracellular and extracellular calcium depletion. Sheared cord platelet Annexin V binding and PAS activity was similar to adult values in GFP, but lower in PRP and WB. Annexin V on sheared cord platelets was calcium-independent, with PAS slightly reduced by intracellular calcium depletion. CONCLUSIONS Increased PS activity on purified sheared cord platelets suggest that their intrinsic function under pathological flow conditions is suppressed by cell-cell or plasmatic components. Although secretory functions of adult and cord platelets retain comparable calcium-dependence, PS exposure in sheared cord platelets is uniquely calcium-independent and distinct from adults. Identification of calcium-regulated developmental disparities in shear-mediated platelet function may provide novel targets for age-specific antiplatelet therapy.
Collapse
Affiliation(s)
- Jawaad Sheriff
- Department of Biomedical Engineering, T08-50 Health Sciences Center, Stony Brook University, Stony Brook, NY 11794-8084 USA
| | - Lisa E. Malone
- Division of Hematology and Oncology, Department of Medicine, Stony Brook University, Stony Brook, NY 11794 USA
| | - Cecilia Avila
- Department of Obstetrics, Gynecology and Reproductive Medicine, Stony Brook University, Stony Brook, NY 11794 USA
| | - Amanda Zigomalas
- Department of Biomedical Engineering, T08-50 Health Sciences Center, Stony Brook University, Stony Brook, NY 11794-8084 USA
| | - Danny Bluestein
- Department of Biomedical Engineering, T08-50 Health Sciences Center, Stony Brook University, Stony Brook, NY 11794-8084 USA
| | - Wadie F. Bahou
- Division of Hematology and Oncology, Department of Medicine, Stony Brook University, Stony Brook, NY 11794 USA
| |
Collapse
|
55
|
Nodeh FK, Hosseini E, Ghasemzadeh M. The effect of gamma irradiation on platelet redox state during storage. Transfusion 2020; 61:579-593. [PMID: 33231307 DOI: 10.1111/trf.16207] [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: 05/30/2020] [Revised: 09/16/2020] [Accepted: 10/28/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND As a method with insignificant adverse effects on in vitro quality of platelet concentrates (PCs), gamma irradiation is applied to abrogate the risk of transfusion-associated graft-vs-host disease in vulnerable recipients. However, there is some evidence of lower posttransfusion responses and proteomic alterations in gamma-irradiated platelets (PLTs), which raises some questions about their quality, safety, and efficacy. Since reactive oxygen species (ROS) are considered as markers of PLT storage lesion (PSL), the study presented here investigated oxidant state in gamma-irradiated PCs. STUDY DESIGN AND METHODS PLT-rich plasma PC was split into two bags, one kept as control while other was subjected to gamma irradiation. Within 7 days of storage, the levels of intra-PLT superoxide, H2 O2 , mitochondrial ROS, P-selectin expression, and phosphatidylserine (PS) exposure were detected by flow cytometry while intracellular reduced glutathione (GSH), glucose concentration, and lactate dehydrogenase (LDH) activity were measured by enzymocolorimetric method. RESULTS GSH decreased, while ROS generation and LDH activity increased, during storage. Gamma irradiation significantly attenuated GSH whereas increased ROS generation in earlier and later stages of storage associated with either P-selectin or PS exposure increments. CONCLUSION Gamma irradiation can significantly increase cytosolic ROS generation in two distinct phases, one upon irradiation and another later in longer-stored PCs. While earlier ROS influx seems to be governed by direct effect of irradiation, the second phase of oxidant stress is presumably due to the storage-dependent PLT activation. Intriguingly, these observations were also in line with early P-selectin increments and increased PS exposure in longer-stored PLTs. Given the mutual link between ROS generation and PLT activation, further investigation is required to explore the effect of gamma irradiation on the induction of PSL.
Collapse
Affiliation(s)
- Fatemeh Kiani Nodeh
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Ehteramolsadat Hosseini
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Mehran Ghasemzadeh
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| |
Collapse
|
56
|
Josefsson EC, Vainchenker W, James C. Regulation of Platelet Production and Life Span: Role of Bcl-xL and Potential Implications for Human Platelet Diseases. Int J Mol Sci 2020; 21:ijms21207591. [PMID: 33066573 PMCID: PMC7589436 DOI: 10.3390/ijms21207591] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/09/2020] [Accepted: 10/10/2020] [Indexed: 01/14/2023] Open
Abstract
Blood platelets have important roles in haemostasis, where they quickly stop bleeding in response to vascular damage. They have also recognised functions in thrombosis, immunity, antimicrobal defense, cancer growth and metastasis, tumour angiogenesis, lymphangiogenesis, inflammatory diseases, wound healing, liver regeneration and neurodegeneration. Their brief life span in circulation is strictly controlled by intrinsic apoptosis, where the prosurvival Bcl-2 family protein, Bcl-xL, has a major role. Blood platelets are produced by large polyploid precursor cells, megakaryocytes, residing mainly in the bone marrow. Together with Mcl-1, Bcl-xL regulates megakaryocyte survival. This review describes megakaryocyte maturation and survival, platelet production, platelet life span and diseases of abnormal platelet number with a focus on the role of Bcl-xL during these processes.
Collapse
Affiliation(s)
- Emma C Josefsson
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC 3052, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, VIC 3052, Australia
| | - William Vainchenker
- University Paris-Saclay, INSERM UMR 1270, Gustave Roussy, 94800 Villejuif, France
| | - Chloe James
- University of Bordeaux, INSERM U1034, Biology of Cardiovascular Diseases, 33600 Pessac, France
- Laboratory of Hematology, Bordeaux University Hospital Center, Haut-Leveque Hospital, 33604 Pessac, France
| |
Collapse
|
57
|
Monzón Manzano E, Fernández-Bello I, Justo Sanz R, Robles Marhuenda Á, López-Longo FJ, Acuña P, Álvarez Román MT, Jiménez Yuste V, Butta NV. Insights into the Procoagulant Profile of Patients with Systemic Lupus Erythematosus without Antiphospholipid Antibodies. J Clin Med 2020; 9:jcm9103297. [PMID: 33066506 PMCID: PMC7602183 DOI: 10.3390/jcm9103297] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/06/2020] [Accepted: 10/11/2020] [Indexed: 12/13/2022] Open
Abstract
We aimed to identify the key players in the prothrombotic profile of patients with systemic lupus erythematosus (SLE) not mediated by antiphospholipid antibodies, as well as the potential utility of global coagulation tests to characterize hemostasis in these patients. Patients with SLE without antiphospholipid antibodies and without signs of thrombosis were included. The kinetics of clot formation were determined by ROTEM®. Platelet activation markers were determined by flow cytometry. Thrombin generation associated with Neutrophil Extracellular Traps (NETs) and microparticles (MPs) was measured by calibrated automated thrombogram (CAT). The plasma levels of PAI-1 were also determined. ROTEM® showed a procoagulant profile in SLE patients. SLE patients had activated platelets and more leukocyte/platelet aggregates at basal conditions. The plasma PAI-1 and platelet aggregates correlated with several ROTEM® parameters. The thrombin generation associated withthe tissue factor (TF) content of MPs and with NETs was increased. Our results suggest the utility of global tests for studying hemostasis in SLE patients because they detect their procoagulant profile, despite having had neither antiphospholipid antibodies nor any previous thrombotic event. A global appraisal of hemostasis should, if possible, be incorporated into clinical practice to detect the risk of a thrombotic event in patients with SLE and to consequently act to prevent its occurrence.
Collapse
Affiliation(s)
- Elena Monzón Manzano
- Hematology Unit, University Hospital La Paz-Idipaz, Paseo de la Castellana 231, 28046 Madrid, Spain; (E.M.M.); (I.F.-B.); (R.J.S.); (P.A.); (M.T.Á.R.); (V.J.Y.)
| | - Ihosvany Fernández-Bello
- Hematology Unit, University Hospital La Paz-Idipaz, Paseo de la Castellana 231, 28046 Madrid, Spain; (E.M.M.); (I.F.-B.); (R.J.S.); (P.A.); (M.T.Á.R.); (V.J.Y.)
| | - Raúl Justo Sanz
- Hematology Unit, University Hospital La Paz-Idipaz, Paseo de la Castellana 231, 28046 Madrid, Spain; (E.M.M.); (I.F.-B.); (R.J.S.); (P.A.); (M.T.Á.R.); (V.J.Y.)
| | | | | | - Paula Acuña
- Hematology Unit, University Hospital La Paz-Idipaz, Paseo de la Castellana 231, 28046 Madrid, Spain; (E.M.M.); (I.F.-B.); (R.J.S.); (P.A.); (M.T.Á.R.); (V.J.Y.)
| | - María Teresa Álvarez Román
- Hematology Unit, University Hospital La Paz-Idipaz, Paseo de la Castellana 231, 28046 Madrid, Spain; (E.M.M.); (I.F.-B.); (R.J.S.); (P.A.); (M.T.Á.R.); (V.J.Y.)
| | - Víctor Jiménez Yuste
- Hematology Unit, University Hospital La Paz-Idipaz, Paseo de la Castellana 231, 28046 Madrid, Spain; (E.M.M.); (I.F.-B.); (R.J.S.); (P.A.); (M.T.Á.R.); (V.J.Y.)
- Faculty of Medicine, Universidad Autónoma de Madrid, 28029 Madrid, Spain
| | - Nora V. Butta
- Hematology Unit, University Hospital La Paz-Idipaz, Paseo de la Castellana 231, 28046 Madrid, Spain; (E.M.M.); (I.F.-B.); (R.J.S.); (P.A.); (M.T.Á.R.); (V.J.Y.)
- Correspondence: ; Tel.: +34-91-727-0000 (ext. 42258)
| |
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
|
Platelets Extracellular Vesicles as Regulators of Cancer Progression-An Updated Perspective. Int J Mol Sci 2020; 21:ijms21155195. [PMID: 32707975 PMCID: PMC7432409 DOI: 10.3390/ijms21155195] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/19/2020] [Accepted: 07/20/2020] [Indexed: 12/14/2022] Open
Abstract
Extracellular vesicles (EVs) are a diverse group of membrane-bound structures secreted in physiological and pathological conditions by prokaryotic and eukaryotic cells. Their role in cell-to-cell communications has been discussed for more than two decades. More attention is paid to assess the impact of EVs in cancer. Numerous papers showed EVs as tumorigenesis regulators, by transferring their cargo molecules (miRNA, DNA, protein, cytokines, receptors, etc.) among cancer cells and cells in the tumor microenvironment. During platelet activation or apoptosis, platelet extracellular vesicles (PEVs) are formed. PEVs present a highly heterogeneous EVs population and are the most abundant EVs group in the circulatory system. The reason for the PEVs heterogeneity are their maternal activators, which is reflected on PEVs size and cargo. As PLTs role in cancer development is well-known, and PEVs are the most numerous EVs in blood, their feasible impact on cancer growth is strongly discussed. PEVs crosstalk could promote proliferation, change tumor microenvironment, favor metastasis formation. In many cases these functions were linked to the transfer into recipient cells specific cargo molecules from PEVs. The article reviews the PEVs biogenesis, cargo molecules, and their impact on the cancer progression.
Collapse
|
60
|
Wolff M, Handtke S, Palankar R, Wesche J, Kohler TP, Kohler C, Gruel Y, Hammerschmidt S, Greinacher A. Activated platelets kill Staphylococcus aureus, but not Streptococcus pneumoniae-The role of FcγRIIa and platelet factor 4/heparinantibodies. J Thromb Haemost 2020; 18:1459-1468. [PMID: 32237268 DOI: 10.1111/jth.14814] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 03/16/2020] [Accepted: 03/23/2020] [Indexed: 02/01/2023]
Abstract
BACKGROUND Heparin induced thrombocytopenia (HIT) is likely a misdirected bacterial host defense mechanism. Platelet factor 4 (PF4) binds to polyanions on bacterial surfaces exposing neo-epitopes to which HIT antibodies bind. Platelets are activated by the resulting immune complexes via FcγRIIA, release bactericidal substances, and kill Gram-negative Escherichia coli. OBJECTIVES To assess the role of PF4, anti-PF4/H antibodies and FcγRIIa in killing of Gram-positive bacteria by platelets. METHODS Binding of PF4 to protein-A deficient Staphylococcus aureus (SA113Δspa) and non-encapsulated Streptococcus pneumoniae (D39Δcps) and its conformational change were assessed by flow cytometry using monoclonal (KKO,5B9) and patient derived anti-PF4/H antibodies. Killing of bacteria was quantified by counting colony forming units (cfu) after incubation with platelets or platelet releasate. Using flow cytometry, platelet activation (CD62P-expression, PAC-1 binding) and phosphatidylserine (PS)-exposure were analyzed. RESULTS Monoclonal and patient-derived anti-PF4/H antibodies bound in the presence of PF4 to both S. aureus and S. pneumoniae (1.6-fold increased fluorescence signal for human anti-PF4/H antibodies to 24.0-fold increase for KKO). Staphylococcus aureus (5.5 × 104 cfu/mL) was efficiently killed by platelets (2.7 × 104 cfu/mL) or their releasate (2.9 × 104 cfu/mL). Killing was not further enhanced by PF4 or anti-PF4/H antibodies. Blocking FcγRIIa had no impact on killing of S. aureus by platelets. In contrast, S. pneumoniae was not killed by platelets or releasate. Instead, after incubation with pneumococci platelets were unresponsive to TRAP-6 stimulation and exposed high levels of PS. CONCLUSIONS Anti-PF4/H antibodies seem to have only a minor role for direct killing of Gram-positive bacteria by platelets. Staphylococcus aureus is killed by platelets or platelet releasate. In contrast, S. pneumoniae affects platelet viability.
Collapse
Affiliation(s)
- Martina Wolff
- Institut für Immunologie und Transfusion Medizin, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Stefan Handtke
- Institut für Immunologie und Transfusion Medizin, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Raghavendra Palankar
- Institut für Immunologie und Transfusion Medizin, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Jan Wesche
- Institut für Immunologie und Transfusion Medizin, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Thomas P Kohler
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, Center for Functional Genomics of Microbes, University of Greifswald, Greifswald, Germany
| | - Christian Kohler
- Institute of Medical Microbiology, University Medicine Greifswald, Greifswald, Germany
| | - Yves Gruel
- Département d'Hématologie-Hémostase, Hôpital Universitaire de Tours, Tours, France
| | - Sven Hammerschmidt
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, Center for Functional Genomics of Microbes, University of Greifswald, Greifswald, Germany
| | - Andreas Greinacher
- Institut für Immunologie und Transfusion Medizin, Universitätsmedizin Greifswald, Greifswald, Germany
| |
Collapse
|
61
|
Waters L, Padula MP, Marks DC, Johnson L. Calcium chelation: a novel approach to reduce cryopreservation-induced damage to frozen platelets. Transfusion 2020; 60:1552-1563. [PMID: 32319689 DOI: 10.1111/trf.15799] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 03/19/2020] [Accepted: 03/20/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Cryopreserved platelets are phenotypically and functionally different to conventionally stored platelets. Calcium may be released from internal stores during the freeze-thaw process, initiating signaling events which lead to these alterations. It was hypothesized that the addition of a calcium chelator prior to cryopreservation may mitigate some of these changes. METHODS Buffy coat-derived platelets that had been pooled and split were tested fresh and following cryopreservation (n = 8 per group). Platelets were cryopreserved using 5%-6% dimethylsulfoxide (DMSO) or were supplemented with increasing concentrations of the internal calcium chelator, BAPTA-AM (100 μM, 200 μM, or 400 μM), prior to storage at -80°C. RESULTS Supplementation of platelets with BAPTA-AM prior to freezing improved platelet recovery in a dose response manner (400 μM: 84 ± 2%) compared to standard DMSO cryopreserved platelets (70 ± 4%). There was a loss of GPIbα, GPVI, and GPIIb/IIIa receptors on platelets following cryopreservation, which was rescued when platelets were supplemented with BAPTA-AM (400 μM: p < 0.0001 for all). Platelet activation markers, such as phosphatidylserine and P-selectin, were externalized on platelets following cryopreservation. However, the addition of BAPTA-AM significantly reduced the increase of these activation markers on cryopreserved platelets (400 μM: p < 0.0001 for both). Both cryopreserved platelet groups exhibited similar functionality as assessed by thromboelastography, forming clots at a faster rate than fresh platelets. CONCLUSIONS This study demonstrates that calcium plays a crucial role in mediating cryopreservation-induced damage to frozen platelets. The addition of the calcium chelator, BAPTA-AM, prior to cryopreservation reduces this damage.
Collapse
Affiliation(s)
- Lauren Waters
- Research and Development, Australian Red Cross Lifeblood (formerly the Australian Red Cross Blood Service), Alexandria, New South Wales, Australia.,School of Life Sciences and Proteomics Core Facility, Faculty of Science, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Matthew P Padula
- School of Life Sciences and Proteomics Core Facility, Faculty of Science, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Denese C Marks
- Research and Development, Australian Red Cross Lifeblood (formerly the Australian Red Cross Blood Service), Alexandria, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Camperdown, New South Wales, Australia
| | - Lacey Johnson
- Research and Development, Australian Red Cross Lifeblood (formerly the Australian Red Cross Blood Service), Alexandria, New South Wales, Australia
| |
Collapse
|
62
|
Śledź KM, Moore SF, Durrant TN, Blair TA, Hunter RW, Hers I. Rapamycin restrains platelet procoagulant responses via FKBP-mediated protection of mitochondrial integrity. Biochem Pharmacol 2020; 177:113975. [PMID: 32298692 DOI: 10.1016/j.bcp.2020.113975] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 04/09/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND PURPOSE Rapamycin is a potent immunosuppressant and anti-proliferative agent used clinically to prevent organ transplant rejection and for coating coronary stents to counteract restenosis. Rapamycin complexes with the immunophilin FKBP12, which subsequently binds and inhibits mTORC1. Despite several reports demonstrating that rapamycin affects platelet-mediated responses, the underlying mechanism of how it alters platelet function is poorly characterised. This study aimed to elucidate the effect of rapamycin on platelet procoagulant responses. EXPERIMENTAL APPROACH The effect of rapamycin on platelet activation and signalling was investigated alongside the catalytic mTOR inhibitors KU0063794 and WYE-687, and the FKBP12-binding macrolide FK506. KEY RESULTS Rapamycin affects platelet procoagulant responses by reducing externalisation of the procoagulant phospholipid phosphatidylserine, formation of balloon-like structures and local generation of thrombin. Catalytic mTOR kinase inhibitors did not alter platelet procoagulant processes, despite having a similar effect as rapamycin on Ca2+ signalling, demonstrating that the effect of rapamycin on procoagulant responses is independent of mTORC1 inhibition and not linked to a reduction in Ca2+ signalling. FK506, which also forms a complex with FKBP12 but does not target mTOR, reduced platelet procoagulant responses to a similar extent as rapamycin. Both rapamycin and FK506 prevented the loss of mitochondria integrity induced by platelet activation, one of the central regulatory events leading to PS externalisation. CONCLUSIONS AND IMPLICATIONS Rapamycin suppresses platelet procoagulant responses by protecting mitochondrial integrity in a manner independent of mTORC1 inhibition. Rapamycin and other drugs targeting FKBP immunophilins could aid the development of novel complementary anti-platelet therapies.
Collapse
Affiliation(s)
- Kamila M Śledź
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, United Kingdom
| | - Samantha F Moore
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, United Kingdom
| | - Tom N Durrant
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, United Kingdom
| | - Thomas A Blair
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, United Kingdom
| | - Roger W Hunter
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, United Kingdom
| | - Ingeborg Hers
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, United Kingdom.
| |
Collapse
|
63
|
Mordakhanova ER, Nevzorova TA, Synbulatova GE, Rauova L, Weisel JW, Litvinov RI. Platelet Activation in Heparin-Induced Thrombocytopenia is Followed by Platelet Death via Complex Apoptotic and Non-Apoptotic Pathways. Int J Mol Sci 2020; 21:ijms21072556. [PMID: 32272655 PMCID: PMC7177543 DOI: 10.3390/ijms21072556] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 04/04/2020] [Accepted: 04/05/2020] [Indexed: 11/16/2022] Open
Abstract
Heparin-induced thrombocytopenia (HIT) is an adverse drug reaction characterized by thrombocytopenia and a high risk for venous or arterial thrombosis. HIT is caused by antibodies that recognize complexes of platelet factor 4 and heparin. The pathogenic mechanisms of this condition are not fully understood. In this study, we used flow cytometry, fluorimetry, and Western blot analysis to study the direct effects of pathogenic immune complexes containing platelet factor 4 on human platelets isolated by gel-filtration. HIT-like pathogenic immune complexes initially caused pronounced activation of platelets detected by an increased expression of phosphatidylserine and P-selectin. This activation was mediated either directly through the FcγRIIA receptors or indirectly via protease-activated receptor 1 (PAR1) receptors due to thrombin generated on or near the surface of activated platelets. The immune activation was later followed by the biochemical signs of cell death, such as mitochondrial membrane depolarization, up-regulation of Bax, down-regulation of Bcl-XL, and moderate activation of procaspase 3 and increased calpain activity. The results show that platelet activation under the action of HIT-like immune complexes is accompanied by their death through complex apoptotic and calpain-dependent non-apoptotic pathways that may underlie the low platelet count in HIT.
Collapse
Affiliation(s)
- Elmira R. Mordakhanova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Tatarstan 420008, Russia; (E.R.M.); (T.A.N.); (G.E.S.)
| | - Tatiana A. Nevzorova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Tatarstan 420008, Russia; (E.R.M.); (T.A.N.); (G.E.S.)
| | - Gulnaz E. Synbulatova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Tatarstan 420008, Russia; (E.R.M.); (T.A.N.); (G.E.S.)
| | - Lubica Rauova
- The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA;
- Departments of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA;
| | - John W. Weisel
- Departments of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA;
| | - Rustem I. Litvinov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Tatarstan 420008, Russia; (E.R.M.); (T.A.N.); (G.E.S.)
- Departments of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
- Correspondence:
| |
Collapse
|
64
|
Naeini MB, Bianconi V, Pirro M, Sahebkar A. The role of phosphatidylserine recognition receptors in multiple biological functions. Cell Mol Biol Lett 2020; 25:23. [PMID: 32226456 PMCID: PMC7098104 DOI: 10.1186/s11658-020-00214-z] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 03/04/2020] [Indexed: 02/06/2023] Open
Abstract
Apoptotic cells are rapidly engulfed and degraded by phagocytes through efferocytosis. Efferocytosis is a highly regulated process. It is triggered upon the activation of caspase-dependent apoptosis, which in turn promotes the expression of "eat me" signals on the surface of dying cells and the release of soluble "find me" signals for the recruitment of phagocytes. To date, many "eat me" signals have been recognized, including phosphatidylserine (PS), intercellular adhesion molecule-3, carbohydrates (e.g., amino sugars, mannose) and calreticulin. Among them, PS is the most studied one. PS recognition receptors are different functionally active receptors expressed by phagocytes. Various PS recognition receptors with different structure, cell type expression, and ability to bind to PS have been recognized. Although PS recognition receptors do not fall into a single classification or family of proteins due to their structural differences, they all share the common ability to activate downstream signaling pathways leading to the production of anti-inflammatory mediators. In this review, available evidence regarding molecular mechanisms underlying PS recognition receptor-regulated clearance of apoptotic cells is discussed. In addition, some efferocytosis-independent biological functions of PS recognition receptors are reviewed.
Collapse
Affiliation(s)
- Mehri Bemani Naeini
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vanessa Bianconi
- Unit of Internal Medicine, Angiology and Arteriosclerosis Diseases, Department of Medicine, University of Perugia, Perugia, Italy
| | - Matteo Pirro
- Unit of Internal Medicine, Angiology and Arteriosclerosis Diseases, Department of Medicine, University of Perugia, Perugia, Italy
| | - Amirhossein Sahebkar
- Halal Research Center of IRI, FDA, Tehran, Iran
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Biotechnology, School of Medicine, Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, P.O. Box: 91779-48564, Mashhad, Iran
| |
Collapse
|
65
|
Calpain cleaves phospholipid flippase ATP8A1 during apoptosis in platelets. Blood Adv 2020; 3:219-229. [PMID: 30674456 DOI: 10.1182/bloodadvances.2018023473] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 12/16/2018] [Indexed: 01/01/2023] Open
Abstract
The asymmetric distribution of phospholipids in the plasma/organellar membranes is generated and maintained through phospholipid flippases in resting cells, but becomes disrupted in apoptotic cells and activated platelets, resulting in phosphatidylserine (PS) exposure on the cell surface. Stable PS exposure during apoptosis requires inactivation of flippases to prevent PS from being reinternalized. Here we show that flippase ATP8A1 is highly expressed in both murine and human platelets, but is not present in the plasma membrane. ATP8A1 is cleaved by the cysteine protease calpain during apoptosis, and the cleavage is prevented indirectly by caspase inhibition, involving blockage of calcium influx into platelets and subsequent calpain activation. In contrast, in platelets activated with thrombin and collagen and exposing PS, ATP8A1 remains intact. These data reveal a novel mechanism of flippase cleavage and suggest that flippase activity in intracellular membranes differs between platelets undergoing apoptosis and activation.
Collapse
|
66
|
Dasgupta SK, Thiagarajan P. Cofilin-1-induced actin reorganization in stored platelets. Transfusion 2020; 60:806-814. [PMID: 32159862 DOI: 10.1111/trf.15747] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 02/13/2020] [Accepted: 02/19/2020] [Indexed: 12/23/2022]
Abstract
BACKGROUND During platelet storage, there are extensive changes in cytoskeleton and phosphatidylserine exposure. The intrinsic mitochondrial pathway of apoptosis, activated in stored platelets, is a major mediator these changes. Cofilin-1 is an effector of actin reorganization. We examined the effect of cofilin-1 deficiency on cytoskeleton and phosphatidylserine exposure during storage and following activation of apoptosis. METHODS AND RESULTS We assessed actin filaments by Alexa-647-phalloidin and phosphatidylserine exposure by fluorescein isothiocyanate-lactadherin by fluorescence microscopy. In fresh platelets, actin filaments are distributed in the subcortical region, and they do not express phosphatidylserine in the outer surface. In stored platelets, there is retraction of actin filaments from the subcortical region with increased phosphatidylserine expression. These changes are seen in 20% of platelets of 6 days old and increases further with storage. Treatment with ABT-737, which activates the mitochondrial apoptosis, induces similar cytoskeletal changes in actin filaments with increased phosphatidylserine. Cofilin-1 is activated in stored platelets as well as in ABT-737 treated platelets by dephosphorylation. In cofilin-1 deficient murine platelets actin filaments are abnormal and ABT-737 induces less phosphatidylserine. Despite these changes in vitro, platelet survival of cofilin-1 deficient platelets in mice was not significantly different from their wild-type controls. CONCLUSION These results show that cofilin-1 plays a role in apoptosis-induced actin rearrangement and phosphatidylserine exposure during storage. Despite the defects in platelet cytoskeleton and phosphatidylserine exposure in cofilin-1-deficient platelets, the in vivo life span of platelets is similar to littermate controls, indicating multiple redundant pathways for the clearance of platelets in vivo.
Collapse
Affiliation(s)
- Swapan K Dasgupta
- Center for Translational Research on Inflammatory Diseases, Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas, USA.,Department of Pathology, Baylor College of Medicine, Houston, Texas, USA
| | - Perumal Thiagarajan
- Center for Translational Research on Inflammatory Diseases, Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas, USA.,Department of Pathology, Baylor College of Medicine, Houston, Texas, USA.,Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
67
|
Reddy EC, Rand ML. Procoagulant Phosphatidylserine-Exposing Platelets in vitro and in vivo. Front Cardiovasc Med 2020; 7:15. [PMID: 32195268 PMCID: PMC7062866 DOI: 10.3389/fcvm.2020.00015] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 01/30/2020] [Indexed: 12/11/2022] Open
Abstract
The physiological heterogeneity of platelets leads to diverse responses and the formation of discrete subpopulations upon platelet stimulation. Procoagulant platelets are an example of such subpopulations, a key characteristic of which is exposure either of the anionic aminophospholipid phosphatidylserine (PS) or of tissue factor on the activated platelet surface. This review focuses on the former, in which PS exposure on a subpopulation of platelets facilitates assembly of the intrinsic tenase and prothrombinase complexes, thereby accelerating thrombin generation on the activated platelet surface, contributing importantly to the hemostatic process. Mechanisms involved in platelet PS exposure, and accompanying events, induced by physiologically relevant agonists are considered then contrasted with PS exposure resulting from intrinsic pathway-mediated apoptosis in platelets. Pathologies of PS exposure, both inherited and acquired, are described. A consideration of platelet PS exposure as an antithrombotic target concludes the review.
Collapse
Affiliation(s)
- Emily C Reddy
- Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON, Canada
| | - Margaret L Rand
- Division of Haematology/Oncology, Translational Medicine, Research Institute, The Hospital for Sick Children, Toronto, ON, Canada.,Departments of Laboratory Medicine & Pathobiology, Biochemistry, and Paediatrics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
68
|
Wang J, Yao Y, Zhang J, Tang X, Meng X, Wang M, Song L, Yuan J. Platelet microRNA-15b protects against high platelet reactivity in patients undergoing percutaneous coronary intervention through Bcl-2-mediated platelet apoptosis. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:364. [PMID: 32355808 PMCID: PMC7186638 DOI: 10.21037/atm.2020.02.88] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Background High platelet reactivity (HPR) and low platelet reactivity (LPR) are associated with an increased risk of ischemic/bleeding events in patients undergoing percutaneous coronary intervention (PCI). The role platelet miRNAs carry out in platelet reactivity regulation is largely unknown. Methods In this study, we profiled the expression pattern of platelet miRNA in patients undergoing PCI with HPR (n=4) and LPR (n=4) by miRNA microarray screening. The candidate miRNAs were further validated in a larger sample of 17 LPR and 22 HPR patients by quantitative reverse-transcription polymerase chain reaction (RT-qPCR), and miR-15b was found differentially expressed. MiR-15b mimic and inhibitor were transfected into MEG-01 cells, then Bcl-2 protein expression and cell apoptosis were assessed. The relationship between platelet reactivity and platelet apoptosis was further evaluated. ABT-737, a Bcl-2 inhibitor was used to induce platelet apoptosis in PCI patients in vitro, and the influence of enhanced platelet apoptosis on platelet reactivity was explored. Results Two miRNAs were found to be differentially expressed in patients with LPR and HPR using microarray system. Furthermore, the expression of miR-15b, a miRNA known to induce cell apoptosis via targeting of Bcl-2, was confirmed by RT-qPCR (P=0.020) to be 1.4× higher in the platelets of LPR patients than in those of HPR patients. Overexpression of miR-15b was demonstrated to suppress Bcl-2 protein expression and enhance cell apoptosis in a megakaryocyte cell line (MEG-01). The platelets of LPR patients expressed lower levels of Bcl-2 protein than those of HPR patients, and an inverse relationship between platelet reactivity and platelet apoptosis was observed among 44 patients who underwent PCI. Inducing platelet apoptosis in PCI patients in vitro, we observed that their platelet reactivity was decreased in a dose-dependent manner. Conclusions Through the promotion of platelet apoptosis, platelet miR-15b negatively regulates platelet reactivity in patients undergoing PCI. Platelet apoptosis may represent a novel antiplatelet target for overcoming HPR in PCI treatment.
Collapse
Affiliation(s)
- Jinghan Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Yi Yao
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Jiahui Zhang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Xiaofang Tang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Xianmin Meng
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Miao Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Lei Song
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Jinqing Yuan
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| |
Collapse
|
69
|
Galle JN, Hegemann JH. Exofacial phospholipids at the plasma membrane: ill-defined targets for early infection processes. Biol Chem 2020; 400:1323-1334. [PMID: 31408428 DOI: 10.1515/hsz-2019-0187] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 08/06/2019] [Indexed: 02/04/2023]
Abstract
The eukaryotic plasma membrane (PM) consists largely of phospholipids and proteins, and separates the intracellular compartments from the extracellular space. It also serves as a signaling platform for cell-to-cell communication and an interaction platform for the molecular crosstalk between pathogens and their target cells. Much research has been done to elucidate the interactions between pathogens and host membrane proteins. However, little is known about the interactions between pathogens and membrane phospholipids, although reports have described a contribution of phospholipids to cell recognition and/or invasion during early infection by diverse pathogens. Thus, during adhesion to the host cell, the obligate intracellular bacterial pathogens Chlamydia spp., the facultative intracellular pathogen Helicobacter pylori and the facultative aerobic pathogen Vibrio parahaemolyticus, interact with exofacial phospholipids. This review focuses on several prominent instances of pathogen interaction with host-cell phospholipids.
Collapse
Affiliation(s)
- Jan N Galle
- Lehrstuhl für Funktionelle Genomforschung der Mikroorganismen, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße 1, D-40225 Düsseldorf, Germany
| | - Johannes H Hegemann
- Lehrstuhl für Funktionelle Genomforschung der Mikroorganismen, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße 1, D-40225 Düsseldorf, Germany
| |
Collapse
|
70
|
Denorme F, Manne BK, Portier I, Eustes AS, Kosaka Y, Kile BT, Rondina MT, Campbell RA. Platelet necrosis mediates ischemic stroke outcome in mice. Blood 2020; 135:429-440. [PMID: 31800959 PMCID: PMC7005363 DOI: 10.1182/blood.2019002124] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 11/15/2019] [Indexed: 12/28/2022] Open
Abstract
Dysregulated platelet functions contribute to the development and progression of ischemic stroke. Utilizing mice with a platelet-specific deletion of cyclophilin D (CypD), a mediator of necrosis, we found that platelet necrosis regulates tissue damage and outcomes during ischemic stroke in vivo. Mice with loss of CypD in platelets (CypDplt-/-mice) exhibited significantly enhanced cerebral blood flow, improved neurological and motor functions, and reduced ischemic stroke infarct volume after cerebral ischemia-reperfusion injury. These effects were attributable, at least in part, to platelet-neutrophil interactions. Twenty-four hours after stroke, significantly more circulating platelet-neutrophil aggregates (PNAs) were found in CypDplt+/+ mice. Underscoring the role of platelet necrosis in PNA formation, we observed a significant number of phosphatidylserine (PS)+ platelets in PNAs in CypDplt+/+ mice. In contrast, significantly fewer platelets in PNAs were PS+ in CypDplt-/- counterparts. Accordingly, mice with CypD-deficient platelets had fewer neutrophils and PNAs recruited to their brain following stroke relative to wild-type counterparts. Neutrophil depletion in wild-type mice conferred protection from ischemic stroke to a similar degree as observed in mice with CypD-deficient platelets. Neutrophil depletion in CypDplt-/- mice did not further reduce infarct size. Transmission electron microscopy of ex vivo-formed PNAs revealed a propensity of necrotic platelets to interact with neutrophils. These results suggest that necrotic platelets interact with neutrophils to exacerbate brain injury during ischemic stroke. Because inhibiting platelet necrosis does not compromise hemostasis, targeting platelet CypD may be a potential therapeutic strategy to limit brain damage following ischemic stroke.
Collapse
Affiliation(s)
- Frederik Denorme
- University of Utah Molecular Medicine Program, Salt Lake City, UT
- Laboratory for Thrombosis Research, Katholieke Universiteit Leuven Campus Kulak Kortrijk, Belgium
| | | | - Irina Portier
- University of Utah Molecular Medicine Program, Salt Lake City, UT
| | - Alicia S Eustes
- University of Utah Molecular Medicine Program, Salt Lake City, UT
| | - Yasuhiro Kosaka
- University of Utah Molecular Medicine Program, Salt Lake City, UT
| | - Benjamin T Kile
- Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Matthew T Rondina
- University of Utah Molecular Medicine Program, Salt Lake City, UT
- Department of Internal Medicine, University of Utah, Salt Lake City, UT
- George E. Wahlen Veterans Affairs Medical Centers Department of Internal Medicine and Geriatric Research Education and Clinical Center, Salt Lake City, UT; and
- Department of Pathology, University of Utah, Salt Lake City, UT
| | - Robert A Campbell
- University of Utah Molecular Medicine Program, Salt Lake City, UT
- Department of Internal Medicine, University of Utah, Salt Lake City, UT
| |
Collapse
|
71
|
Stolla M, Bailey SL, Fang L, Fitzpatrick L, Gettinger I, Pellham E, Christoffel T. Effects of storage time prolongation on in vivo and in vitro characteristics of 4°C-stored platelets. Transfusion 2020; 60:613-621. [PMID: 32017135 DOI: 10.1111/trf.15669] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 11/30/2019] [Accepted: 12/09/2019] [Indexed: 12/28/2022]
Abstract
BACKGROUND Cold (4°C)-stored platelets are currently under investigation for transfusion in bleeding patients. It is currently unknown how long cold-stored platelets can be stored for clinical applications. STUDY DESIGN AND METHODS Twenty three subjects were recruited. Twenty-one subjects were available for in vivo assessment and received indium-111 radiolabeled, cold-stored platelets. We investigated 5- (n = 5), 10- (n = 6), 15- (n = 5), and 20-day-stored (n = 5) platelets and obtained samples for in vitro testing at baseline and after the designated storage time. Twenty three units were available for in vitro testing. Five- and 7-day (n = 5 each), room temperature (RT)-stored platelets served as the current clinical standard control. RESULTS In vivo, we found a continuous decline in platelet recovery from 5 to 20 days. Platelet survival reached a low nadir after 10 days of storage. Ex vivo, we observed the maximum platelet αIIbβ3 integrin response to collagen at 5 days of cold storage, and we saw a continuous decline thereafter. However, platelet integrin activation and mitochondrial membrane integrity were better preserved after 20 days at 4°C, compared to 5 days at RT. Platelet metabolic parameters suggest comparable results between 20-day cold-stored platelets and 5- or 7-day RT-stored platelets. CONCLUSION In summary, we performed the first studies with extended, cold-stored, apheresis platelets in plasma for up to 20 days with a fresh comparator. Storing cold-stored platelets up to 20 days yields better results in vitro, but further studies in actively bleeding patients are needed to determine the best compromise between hemostatic efficacy and storage prolongation.
Collapse
Affiliation(s)
- Moritz Stolla
- Platelet Transfusion Research Laboratory, Bloodworks Northwest Research Institute, Seattle, Washington.,Department of Medicine, Division of Hematology, University of Washington School of Medicine, Seattle, Washington
| | - S Lawrence Bailey
- Platelet Transfusion Research Laboratory, Bloodworks Northwest Research Institute, Seattle, Washington
| | - Lydia Fang
- Platelet Transfusion Research Laboratory, Bloodworks Northwest Research Institute, Seattle, Washington
| | - Lynda Fitzpatrick
- Platelet Transfusion Research Laboratory, Bloodworks Northwest Research Institute, Seattle, Washington
| | - Irena Gettinger
- Platelet Transfusion Research Laboratory, Bloodworks Northwest Research Institute, Seattle, Washington
| | - Esther Pellham
- Platelet Transfusion Research Laboratory, Bloodworks Northwest Research Institute, Seattle, Washington
| | - Todd Christoffel
- Platelet Transfusion Research Laboratory, Bloodworks Northwest Research Institute, Seattle, Washington
| |
Collapse
|
72
|
Hewes JL, Lee JY, Fagan KA, Bauer NN. The changing face of pulmonary hypertension diagnosis: a historical perspective on the influence of diagnostics and biomarkers. Pulm Circ 2020; 10:2045894019892801. [PMID: 32110383 PMCID: PMC7000867 DOI: 10.1177/2045894019892801] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 11/11/2019] [Indexed: 12/15/2022] Open
Abstract
Pulmonary hypertension is a complex, multifactorial disease that results in right heart failure and premature death. Since the initial reports of pulmonary hypertension in the late 1800s, the diagnosis of pulmonary hypertension has evolved with respect to its definition, screening tools, and diagnostic techniques. This historical perspective traces the earliest roots of pulmonary hypertension detection and diagnosis through to the current recommendations for classification. We highlight the diagnostic tools used in the past and present, and end with a focus on the future directions of early detection. Early detection of pulmonary hypertension and pulmonary arterial hypertension and the proper determination of etiology are vital for the early therapeutic intervention that can prolong life expectancy and improve quality of life. The search for a non-invasive screening tool for the identification and classification of pulmonary hypertension is ongoing, and we discuss the role of animal models of the disease in this search.
Collapse
Affiliation(s)
- Jenny L. Hewes
- Department of Pharmacology, College of
Medicine,
University
of South Alabama, Mobile, AL, USA
- Center for Lung Biology, College of
Medicine,
University
of South Alabama, Mobile, AL, USA
| | - Ji Young Lee
- Center for Lung Biology, College of
Medicine,
University
of South Alabama, Mobile, AL, USA
- Division of Pulmonary and Critical Care
Medicine, University Hospital,
University
of South Alabama, Mobile, AL, USA
- Department of Physiology and Cell
Biology, College of Medicine,
University
of South Alabama, Mobile, AL, USA
| | - Karen A. Fagan
- Department of Pharmacology, College of
Medicine,
University
of South Alabama, Mobile, AL, USA
- Center for Lung Biology, College of
Medicine,
University
of South Alabama, Mobile, AL, USA
- Division of Pulmonary and Critical Care
Medicine, University Hospital,
University
of South Alabama, Mobile, AL, USA
| | - Natalie N. Bauer
- Department of Pharmacology, College of
Medicine,
University
of South Alabama, Mobile, AL, USA
- Center for Lung Biology, College of
Medicine,
University
of South Alabama, Mobile, AL, USA
| |
Collapse
|
73
|
Pennell EN, Shiels R, Vidimce J, Wagner KH, Shibeeb S, Bulmer AC. The impact of bilirubin ditaurate on platelet quality during storage. Platelets 2019; 31:884-896. [PMID: 31747815 DOI: 10.1080/09537104.2019.1693038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Bilirubin ditaurate (BRT), a conjugated bilirubin analogue, has demonstrated anti-platelet characteristics following acute ex vivo exposure. Scavenging of mitochondrial superoxide and attenuation of granule exocytosis suggested a potential benefit for including BRT for storage. With no reports of cytotoxicity following acute exposure, the impact of 35µM BRT on platelet function was investigated, in clinically suppled units, for up to seven days. Exposure to 35µM BRT significantly reduced mitochondrial membrane potential and increased glucose consumption until exhaustion after 72 hours. Platelet aggregation and activation was significantly impaired by BRT. Mitochondrial superoxide production and phosphatidylserine expression were significantly elevated following glucose exhaustion, with decreased viability observed from day five onwards. Lactate accumulation and loss of bicarbonate, support a metabolic disturbance, leading to a decline of quality following BRT inclusion. Although acute ex vivo BRT exposure reported potentially beneficial effects, translation from acute to chronic exposure failed to combat declining platelet function during storage. BRT exposure resulted in perturbations of platelet quality, with the utility of BRT during storage therefore limited. However, these are the first data of prolonged platelet exposure to analogues of conjugated bilirubin and may improve our understanding of platelet function in the context of conjugated hyperbilirubinemia.
Collapse
Affiliation(s)
- Evan Noel Pennell
- School of Medical Science, Griffith University , Gold Coast, Australia
| | - Ryan Shiels
- School of Medical Science, Griffith University , Gold Coast, Australia
| | - Josif Vidimce
- School of Medical Science, Griffith University , Gold Coast, Australia
| | - Karl-Heinz Wagner
- Research Platform Active Aging, Department of Nutritional Science, University of Vienna , Vienna Austria
| | - Sapha Shibeeb
- School of Medical Science, Griffith University , Gold Coast, Australia.,Endeavour College of Natural Health , Melbourne, Australia
| | | |
Collapse
|
74
|
Platelet CD36 signaling through ERK5 promotes caspase-dependent procoagulant activity and fibrin deposition in vivo. Blood Adv 2019; 2:2848-2861. [PMID: 30381401 DOI: 10.1182/bloodadvances.2018025411] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 09/28/2018] [Indexed: 12/12/2022] Open
Abstract
Dyslipidemia is a risk factor for clinically significant thrombotic events. In this condition, scavenger receptor CD36 potentiates platelet reactivity through recognition of circulating oxidized lipids. CD36 promotes thrombosis by activating redox-sensitive signaling molecules, such as the MAPK extracellular signal-regulated kinase 5 (ERK5). However, the events downstream of platelet ERK5 are not clear. In this study, we report that oxidized low-density lipoprotein (oxLDL) promotes exposure of procoagulant phosphatidylserine (PSer) on platelet surfaces. Studies using pharmacologic inhibitors indicate that oxLDL-CD36 interaction-induced PSer exposure requires apoptotic caspases in addition to the downstream CD36-signaling molecules Src kinases, hydrogen peroxide, and ERK5. Caspases promote PSer exposure and, subsequently, recruitment of the prothrombinase complex, resulting in the generation of fibrin from the activation of thrombin. Caspase activity was observed when platelets were stimulated with oxLDL. This was prevented by inhibiting CD36 and ERK5. Furthermore, oxLDL potentiates convulxin/glycoprotein VI-mediated fibrin formation by platelets, which was prevented when CD36, ERK5, and caspases were inhibited. Using 2 in vivo arterial thrombosis models in apoE-null hyperlipidemic mice demonstrated enhanced arterial fibrin accumulation upon vessel injury. Importantly, absence of ERK5 in platelets or mice lacking CD36 displayed decreased fibrin accumulation in high-fat diet-fed conditions comparable to that seen in chow diet-fed animals. These findings suggest that platelet signaling through CD36 and ERK5 induces a procoagulant phenotype in the hyperlipidemic environment by enhancing caspase-mediated PSer exposure.
Collapse
|
75
|
Yang M, Silverstein RL. CD36 and ERK5 link dyslipidemia to apoptotic-like platelet procoagulant function. Curr Opin Hematol 2019; 26:357-365. [PMID: 31261174 PMCID: PMC9308374 DOI: 10.1097/moh.0000000000000522] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
PURPOSE OF REVIEW Metabolic diseases, including dyslipidemia, diabetes mellitus, and chronic inflammation are risk factors for clinically significant thrombotic events. Thrombosis in these settings is multifaceted with coordinated mechanisms between platelet activation and the hemostatic pathways. This review focuses on recent advances in platelet procoagulant and apoptotic signaling with emphasis on the pathophysiologic mechanisms induced by platelet CD36 in dyslipidemia, and the key unaddressed questions relating to the field. RECENT FINDINGS CD36 promotes platelet activation and increases the risk for thrombosis through signaling events. These include generation of reactive oxygen species, activation of redox-sensitive MAP kinase ERK5, and promotion of a pro-thrombotic phenotype. CD36 promotes phosphatidylserine externalization leading to a procoagulant function downstream from MAP kinase ERK5 that is separate from a pro-aggregatory function. Phosphatidylserine externalization requires maladaptive caspase activation, promotes assembly of the factor tenase and prothrombinase complex, and promotes fibrin formation. It is distinct from the canonical pathways mediating platelet procoagulant function by strong physiologic stimuli or by the platelet apoptotic-like Bak/Bax-mediated pathway for cellular clearance. SUMMARY Understanding CD36 signaling in the context of dyslipidemia, or other metabolic diseases will identify important and novel signaling hubs that could be potential therapeutic targets for intervention without impacting hemostasis.
Collapse
Affiliation(s)
- Moua Yang
- Department of Biochemistry, Medical College of Wisconsin
- Blood Research Institute, Versiti Blood Center of Wisconsin
| | - Roy L. Silverstein
- Blood Research Institute, Versiti Blood Center of Wisconsin
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
76
|
Fuentes E, Araya-Maturana R, Urra FA. Regulation of mitochondrial function as a promising target in platelet activation-related diseases. Free Radic Biol Med 2019; 136:172-182. [PMID: 30625393 DOI: 10.1016/j.freeradbiomed.2019.01.007] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 12/22/2018] [Accepted: 01/04/2019] [Indexed: 12/13/2022]
Abstract
Platelets are anucleated cell elements produced by fragmentation of the cytoplasm of megakaryocytes and have a unique metabolic phenotype compared with circulating leukocytes, exhibiting a high coupling efficiency to mitochondrial adenosine triphosphate production with reduced respiratory reserve capacity. Platelet mitochondria are well suited for ex vivo analysis of different diseases. Even some diseases induce mitochondrial changes in platelets without reflecting them in other organs. During platelet activation, an integrated participation of glycolysis and oxidative phosphorylation is mediated by oxidative stress production-dependent signaling. The platelet activation-dependent procoagulant activity mediated by collagen, thrombin and hyperglycemia induce mitochondrial dysfunction to promote thrombosis in oxidative stress-associated pathological conditions. Interestingly, some compounds exhibit a protective action on platelet mitochondrial dysfunction through control of mitochondrial oxidative stress production or inhibition of respiratory complexes. They can be grouped in a) Natural source-derived compounds (e.g. Xanthohumol, Salvianoloc acid A and Sila-amide derivatives of NAC), b) TPP+-linked small molecules (e.g. mitoTEMPO and mitoQuinone) and c) FDA-approved drugs (e.g. metformin and statins), illustrating the wide range of molecular structures capable of effectively interacting with platelet mitochondria. The present review article aims to discuss the mechanisms of mitochondrial dysfunction and their association with platelet activation-related diseases.
Collapse
Affiliation(s)
- Eduardo Fuentes
- Thrombosis Research Center, Medical Technology School, Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Interdisciplinary Excellence Research Program on Healthy Aging (PIEI-ES), Universidad de Talca, Talca, Chile.
| | - Ramiro Araya-Maturana
- Instituto de Química de Recursos Naturales, Programa de Investigación Asociativa en Cáncer Gástrico (PIA-CG), Universidad de Talca, Talca, Chile
| | - Félix A Urra
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile.
| |
Collapse
|
77
|
Nurden AT. Acquired Glanzmann thrombasthenia: From antibodies to anti-platelet drugs. Blood Rev 2019; 36:10-22. [PMID: 31010659 DOI: 10.1016/j.blre.2019.03.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 03/15/2019] [Accepted: 03/19/2019] [Indexed: 02/06/2023]
Abstract
In contrast to the inherited platelet disorder given by mutations in the ITGA2B and ITGB3 genes, mucocutaneous bleeding from a spontaneous inhibition of normally expressed αIIbβ3 characterizes acquired Glanzmann thrombasthenia (GT). Classically, it is associated with autoantibodies or paraproteins that block platelet aggregation without causing a fall in platelet count. However, inhibitory antibodies to αIIbβ3 are widely associated with primary immune thrombocytopenia (ITP), occur in secondary ITP associated with leukemia and related disorders, solid cancers and myeloma, other autoimmune diseases, following organ transplantation while cytoplasmic dysregulation of αIIbβ3 function features in myeloproliferative and myelodysplastic syndromes. Antibodies to αIIbβ3 occur during viral and bacterial infections, while drug-dependent antibodies reacting with αIIbβ3 are a special case. Direct induction of acquired GT is a feature of therapies that block platelets in coronary artery disease. This review looks at these conditions, emphasizing molecular mechanisms, therapy, patient management and future directions for research.
Collapse
Affiliation(s)
- Alan T Nurden
- Institut de Rhythmologie et de Modélisation Cardiaque, Plateforme Technologique d'Innovation Biomédicale, Hôpital Xavier Arnozan, Pessac, France.
| |
Collapse
|
78
|
|
79
|
Boulet C, Doerig CD, Carvalho TG. Manipulating Eryptosis of Human Red Blood Cells: A Novel Antimalarial Strategy? Front Cell Infect Microbiol 2018; 8:419. [PMID: 30560094 PMCID: PMC6284368 DOI: 10.3389/fcimb.2018.00419] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 11/14/2018] [Indexed: 12/13/2022] Open
Abstract
Malaria is a major global health burden, affecting over 200 million people worldwide. Resistance against all currently available antimalarial drugs is a growing threat, and represents a major and long-standing obstacle to malaria eradication. Like many intracellular pathogens, Plasmodium parasites manipulate host cell signaling pathways, in particular programmed cell death pathways. Interference with apoptotic pathways by malaria parasites is documented in the mosquito and human liver stages of infection, but little is known about this phenomenon in the erythrocytic stages. Although mature erythrocytes have lost all organelles, they display a form of programmed cell death termed eryptosis. Numerous features of eryptosis resemble those of nucleated cell apoptosis, including surface exposure of phosphatidylserine, cell shrinkage and membrane ruffling. Upon invasion, Plasmodium parasites induce significant stress to the host erythrocyte, while delaying the onset of eryptosis. Many eryptotic inducers appear to have a beneficial effect on the course of malaria infection in murine models, but major gaps remain in our understanding of the underlying molecular mechanisms. All currently available antimalarial drugs have parasite-encoded targets, which facilitates the emergence of resistance through selection of mutations that prevent drug-target binding. Identifying host cell factors that play a key role in parasite survival will provide new perspectives for host-directed anti-malarial chemotherapy. This review focuses on the interrelationship between Plasmodium falciparum and the eryptosis of its host erythrocyte. We summarize the current knowledge in this area, highlight the different schools of thoughts and existing gaps in knowledge, and discuss future perspectives for host-directed therapies in the context of antimalarial drug discovery.
Collapse
Affiliation(s)
- Coralie Boulet
- Molecular Parasitology Laboratory, Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, Australia
| | - Christian D Doerig
- Infection and Immunity Program, Biomedicine Discovery Institute, Department of Microbiology, Monash University, Clayton, VIC, Australia
| | - Teresa G Carvalho
- Molecular Parasitology Laboratory, Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, Australia
| |
Collapse
|
80
|
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
|
81
|
Kholmukhamedov A, Janecke R, Choo HJ, Jobe SM. The mitochondrial calcium uniporter regulates procoagulant platelet formation. J Thromb Haemost 2018; 16:2315-2321. [PMID: 30179298 DOI: 10.1111/jth.14284] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Indexed: 12/01/2022]
Abstract
Essentials Mitochondrial hyperpolarization enhances the conversion of platelets to a procoagulant phenotype. Mitochondrial calcium uniporter (MCU) function is essential in procoagulant platelet formation. Mitochondrial calcium uniporter deletion does not impact other aspects of platelet activation. Ablation of MCU results in the emergence of a permeability transition pore-independent pathway. SUMMARY: Background Procoagulant platelets comprise a phenotypically distinct subpopulation of activated platelets with high-level phosphatidylserine externalization. When initiated by co-stimulation with thrombin and a glycoprotein VI (GPVI) agonist, the transition to the procoagulant phenotype is mediated by extracellular calcium entry and mitochondrial permeability transition pore (mPTP) formation. Objectives The intracellular mechanisms coordinating these distinct cytoplasmic and mitochondrial processes remain unclear. The mitochondrial calcium uniporter (MCU) protein is a central component of the transmembrane ion channel that allows the passage of Ca2+ from the cytosol into the mitochondrial matrix. Here we investigate the role of the MCU in the regulation of procoagulant platelet formation. Results Procoagulant platelet formation was directly correlated with pre-stimulatory mitochondrial transmembrane potential, a key determinant of calcium flux from the cytoplasm to the mitochondria. The role of MCU in the regulation of procoagulant platelet formation was investigated using MCU null platelets. Procoagulant platelet formation in MCU null platelets was significantly decreased coincident with decreased mPTP formation. In contrast, neither granule release nor initial integrin activation was altered in response to stimulation. In the genomic absence of MCU, developmental induction of an alternative intracellular pathway partially rescued procoagulant platelet formation. Conclusion These results identify a key role for the mitochondrial calcium uptake channel in the regulation of mPTP-mediated procoagulant platelet formation and suggest a novel pharmacologic target for procoagulant-platelet-related pathologies.
Collapse
Affiliation(s)
| | - R Janecke
- Blood Center of Wisconsin, Milwaukee, WI, USA
| | - H-J Choo
- Emory University, Atlanta, GA, USA
| | - S M Jobe
- Blood Center of Wisconsin, Milwaukee, WI, USA
- Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
82
|
Rath D, Chatterjee M, Meyer L, Tekath N, Olma C, Krumm P, Adams C, Borst O, Müller K, Droppa M, Nikolaou K, Riethmüller J, Gawaz M, Geisler T. Relative survival potential of platelets is associated with platelet CXCR4/CXCR7 surface exposure and functional recovery following STEMI. Atherosclerosis 2018; 278:269-277. [PMID: 30342381 DOI: 10.1016/j.atherosclerosis.2018.10.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 09/16/2018] [Accepted: 10/05/2018] [Indexed: 10/28/2022]
Abstract
BACKGROUND AND AIMS Platelets are critically involved in tissue repair and regeneration, which depend on their inflammatory properties and survival. SDF-1 ligates to CXCR4 and CXCR7 and contributes to the regulation of platelet survival. Platelet CXCR4/CXCR7 are involved in myocardial regeneration after infarction and are associated with outcomes in patients with symptomatic coronary artery disease. This study investigates the CXCR4/CXCR7 platelet survival axis ex vivo. METHODS 87 patients with ST-segment elevation myocardial infarction (STEMI) were included and analyzed for platelet surface exposure of CXCR4, CXCR7, Annexin V binding and tetramethylrhodamine ethyl ester (TMRE) response. Serum of 38 patients was analyzed for FasL, TNFα, TNF RI, TNF RII and TRAIL with Bioplex®. The majority of patients received sequential cardiac MRI (intrahospital, 6-month follow-up). RESULTS We found a strong and positive correlation between surface exposure of CXCR4 and CXCR7 (ρ = 0.856, p<0.001). Relative survival potential correlated significantly with both platelet surface exposure of CXCR4 and CXCR7 (ρ = 0.365, p = 0.019; ρ = 0.417, p = 0.006) and furthermore with improvement of myocardial left ventricular ejection fraction (LVEF) (ρ = 0.490, p = 0.013). High relative survival potential showed significantly higher levels for both CXCR4 and CXCR7 surface exposure (MFI 87.3 vs. 69.0, p = 0.037; MFI 71.4 vs. 59.3, p = 0.045). We found a significant change in absolute LVEF% over the course of 6 months in patients with high CXCR7 platelet surface exposure (LVEF% 44.3 vs. 60.0, p≤0.001). CONCLUSIONS Platelet survival is associated with platelet surface exposure of CXCR4 and CXCR7 in STEMI patients and contributes to functional recovery after STEMI.
Collapse
Affiliation(s)
- Dominik Rath
- Department of Internal Medicine III, Cardiology and Cardiovascular Medicine, University of Tuebingen, Germany
| | - Madhumita Chatterjee
- Department of Internal Medicine III, Cardiology and Cardiovascular Medicine, University of Tuebingen, Germany
| | - Lennart Meyer
- Department of Internal Medicine III, Cardiology and Cardiovascular Medicine, University of Tuebingen, Germany
| | - Nina Tekath
- Department of Internal Medicine III, Cardiology and Cardiovascular Medicine, University of Tuebingen, Germany
| | - Carolin Olma
- Department of Internal Medicine III, Cardiology and Cardiovascular Medicine, University of Tuebingen, Germany
| | - Patrick Krumm
- Department of Radiology, Diagnostic and Interventional Radiology, University of Tuebingen, Germany
| | - Constantin Adams
- Center for Pediatric Clinical Studies, University of Tuebingen, Germany
| | - Oliver Borst
- Department of Internal Medicine III, Cardiology and Cardiovascular Medicine, University of Tuebingen, Germany
| | - Karin Müller
- Department of Internal Medicine III, Cardiology and Cardiovascular Medicine, University of Tuebingen, Germany
| | - Michal Droppa
- Department of Internal Medicine III, Cardiology and Cardiovascular Medicine, University of Tuebingen, Germany
| | - Konstantin Nikolaou
- Department of Radiology, Diagnostic and Interventional Radiology, University of Tuebingen, Germany
| | | | - Meinrad Gawaz
- Department of Internal Medicine III, Cardiology and Cardiovascular Medicine, University of Tuebingen, Germany
| | - Tobias Geisler
- Department of Internal Medicine III, Cardiology and Cardiovascular Medicine, University of Tuebingen, Germany.
| |
Collapse
|
83
|
Loss of the mitochondrial kinase PINK1 does not alter platelet function. Sci Rep 2018; 8:14377. [PMID: 30258205 PMCID: PMC6158262 DOI: 10.1038/s41598-018-32716-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 09/07/2018] [Indexed: 11/24/2022] Open
Abstract
PTEN-induced putative kinase (PINK) 1 is regarded as a master regulator of cellular mitophagy such that loss of function mutations contribute to early onset Parkinson’s disease, through aberrant mitochondrial control and function. Mitochondrial function is key to platelet procoagulant activity, controlling the haemostatic response to vessel injury, but can also predispose blood vessels to thrombotic complications. Here, we sought to determine the role of PINK1 in platelet mitochondrial health and function using PINK1 knockout (KO) mice. The data largely show an absence of such a role. Haematological analysis of blood counts from KO mice was comparable to wild type. Quantification of mitochondrial mass by citrate synthase activity assay or expression of mitochondrial markers were comparable, suggesting normal mitophagy in KO platelets. Analysis of mitochondrial permeability transition pore opening, changes in mitochondrial membrane potential and calcium signalling to platelet activation were unaffected by loss of PINK1, whereas subtle enhancements of activation-induced reactive oxygen species were detected. Platelet aggregation, integrin activation, α- and dense granule secretion and phosphatidylserine exposure were unaltered in KO platelets while mouse tail bleeding responses were similar to wild type. Together these results demonstrate that PINK1 does not regulate basal platelet mitophagy and is dispensable for platelet function.
Collapse
|
84
|
Pang A, Cui Y, Chen Y, Cheng N, Delaney MK, Gu M, Stojanovic-Terpo A, Zhu C, Du X. Shear-induced integrin signaling in platelet phosphatidylserine exposure, microvesicle release, and coagulation. Blood 2018; 132:533-543. [PMID: 29853537 PMCID: PMC6073322 DOI: 10.1182/blood-2017-05-785253] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 05/22/2018] [Indexed: 01/07/2023] Open
Abstract
It is currently unclear why agonist-stimulated platelets require shear force to efficiently externalize the procoagulant phospholipid phosphatidylserine (PS) and release PS-exposed microvesicles (MVs). We reveal that integrin outside-in signaling is an important mechanism for this requirement. PS exposure and MV release were inhibited in β3-/- platelets or by integrin antagonists. The impaired MV release and PS exposure in β3-/- platelets were rescued by expression of wild-type β3 but not a Gα13 binding-deficient β3 mutant (E733EE to AAA), which blocks outside-in signaling but not ligand binding. Inhibition of Gα13 or Src also diminished agonist/shear-dependent PS exposure and MV release, further indicating a role for integrin outside-in signaling. PS exposure in activated platelets was induced by application of pulling force via an integrin ligand, which was abolished by inhibiting Gα13-integrin interaction, suggesting that Gα13-dependent transmission of mechanical signals by integrins induces PS exposure. Inhibition of Gα13 delayed coagulation in vitro. Furthermore, inhibition or platelet-specific knockout of Gα13 diminished laser-induced intravascular fibrin formation in arterioles in vivo. Thus, β3 integrins serve as a shear sensor activating the Gα13-dependent outside-in signaling pathway to facilitate platelet procoagulant function. Pharmacological targeting of Gα13-integrin interaction prevents occlusive thrombosis in vivo by inhibiting both coagulation and platelet thrombus formation.
Collapse
Affiliation(s)
- Aiming Pang
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL
- Hematopoietic Stem Cell Transplantation Center, Institute of Hematology and Blood Diseases Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Tianjin, China
| | - Yujie Cui
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL
- School of Medical Laboratory, Tianjin Medical University, Tianjin, China
| | - Yunfeng Chen
- Woodruff School of Mechanical Engineering and
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA
| | - Ni Cheng
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL
| | - M Keegan Delaney
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL
- Dupage Medical Technology, Inc., Willowbrook, IL; and
| | - Minyi Gu
- Dupage Medical Technology, Inc., Willowbrook, IL; and
| | | | - Cheng Zhu
- Woodruff School of Mechanical Engineering and
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA
| | - Xiaoping Du
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL
| |
Collapse
|
85
|
Qasim H, Karim ZA, Silva-Espinoza JC, Khasawneh FT, Rivera JO, Ellis CC, Bauer SL, Almeida IC, Alshbool FZ. Short-Term E-Cigarette Exposure Increases the Risk of Thrombogenesis and Enhances Platelet Function in Mice. J Am Heart Assoc 2018; 7:JAHA.118.009264. [PMID: 30021806 PMCID: PMC6201451 DOI: 10.1161/jaha.118.009264] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Background Cardiovascular disease is the main cause of death in the United States, with smoking being the primary preventable cause of premature death, and thrombosis being the main mechanism of cardiovascular mortality in smokers. Due to the perception that electronic/e‐cigarettes are “safer/less harmful” than conventional cigarettes, their usage—among a variety of ages—has increased tremendously during the past decade. Notably, there are limited studies regarding the negative effects of e‐cigarettes on the cardiovascular system, which is also the subject of significant debate. Methods and Results We employed a passive e‐VapeTM vapor inhalation system and developed an in vivo whole‐body e‐cigarette mouse exposure protocol that mimics real‐life human exposure scenarios/conditions and investigated the effects of e‐cigarettes and clean air on platelet function and thrombogenesis. Our results show that platelets from e‐cigarette–exposed mice are hyperactive, with enhanced aggregation, dense and α granule secretion, activation of the αIIbβ3 integrin, phosphatidylserine expression, and Akt and ERK activation, when compared with clean air–exposed platelets. E‐cigarette–exposed platelets were also found to be resistant to inhibition by prostacyclin, relative to clean air. Furthermore, the e‐cigarette–exposed mice exhibited a shortened thrombosis occlusion and bleeding times. Conclusions Taken together, our data demonstrate for the first time that e‐cigarettes alter physiological hemostasis and increase the risk of thrombogenic events. This is attributable, at least in part, to the hyperactive state of platelets. Thus, the negative health consequences of e‐cigarette exposure should not be underestimated and warrant further investigation.
Collapse
Affiliation(s)
- Hanan Qasim
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas El Paso, TX
| | - Zubair A Karim
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas El Paso, TX
| | - Juan C Silva-Espinoza
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas El Paso, TX
| | - Fadi T Khasawneh
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas El Paso, TX
| | - José O Rivera
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas El Paso, TX
| | - Cameron C Ellis
- Border Biomedical Research Center, Department of Biological Sciences, College of Science, University of Texas El Paso, TX
| | - Stephanie L Bauer
- Border Biomedical Research Center, Department of Biological Sciences, College of Science, University of Texas El Paso, TX
| | - Igor C Almeida
- Border Biomedical Research Center, Department of Biological Sciences, College of Science, University of Texas El Paso, TX
| | - Fatima Z Alshbool
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas El Paso, TX
| |
Collapse
|
86
|
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
|
87
|
Swieringa F, Spronk HM, Heemskerk JW, van der Meijden PE. Integrating platelet and coagulation activation in fibrin clot formation. Res Pract Thromb Haemost 2018; 2:450-460. [PMID: 30046749 PMCID: PMC6046596 DOI: 10.1002/rth2.12107] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 03/31/2018] [Indexed: 12/21/2022] Open
Abstract
Platelets interact with the coagulation system in a multitude of ways, not only during the phases of thrombus formation, but also in specific areas within a formed thrombus. This review discusses current concepts of platelet control of thrombin generation, fibrin formation and structure, and anticoagulation. Indicated are how combined signalling via the platelet receptors for collagen (glycoprotein VI) and thrombin induces the secretion of (anti)coagulation factors, as well as surface exposure of phosphatidylserine, thereby catalysing thrombin generation. This procoagulant platelet response is also facilitated by the adhesive complexes glycoprotein Ib-V-IX and integrin αIIbβ3. In the buildup of a platelet-fibrin thrombus, the extrinsic, tissue factor-driven coagulation pathway is predominant in early stages, while the intrinsic, factor XII pathway seems to promote at later time points. Already early generation of thrombin enforces platelet responses and stimulates intra-thrombus heterogeneity with patches of loosely aggregated, contracted, and phosphatidylserine-exposing platelets. Fibrin actively formed on the surface of activated platelets supports thrombus growth, but also captures thrombin. The fibrin distribution in a thrombus appears to rely on the local procoagulant trigger and the blood flow rate. Clinical studies support the importance of the platelet-coagulation interplay, by showing beneficial effects of combination therapy in the secondary prevention of cardiovascular disease.
Collapse
Affiliation(s)
- Frauke Swieringa
- Department of BiochemistryCardiovascular Research Institute Maastricht (CARIM)Maastricht UniversityMaastrichtThe Netherlands
- Leibniz Institute for Analytical SciencesISASDortmundGermany
| | - Henri M.H. Spronk
- Department of BiochemistryCardiovascular Research Institute Maastricht (CARIM)Maastricht UniversityMaastrichtThe Netherlands
| | - Johan W.M. Heemskerk
- Department of BiochemistryCardiovascular Research Institute Maastricht (CARIM)Maastricht UniversityMaastrichtThe Netherlands
| | - Paola E.J. van der Meijden
- Department of BiochemistryCardiovascular Research Institute Maastricht (CARIM)Maastricht UniversityMaastrichtThe Netherlands
| |
Collapse
|
88
|
Fidler TP, Campbell RA, Funari T, Dunne N, Balderas Angeles E, Middleton EA, Chaudhuri D, Weyrich AS, Abel ED. Deletion of GLUT1 and GLUT3 Reveals Multiple Roles for Glucose Metabolism in Platelet and Megakaryocyte Function. Cell Rep 2018; 20:881-894. [PMID: 28746873 DOI: 10.1016/j.celrep.2017.06.083] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 05/08/2017] [Accepted: 06/27/2017] [Indexed: 01/08/2023] Open
Abstract
Anucleate platelets circulate in the blood to facilitate thrombosis and diverse immune functions. Platelet activation leading to clot formation correlates with increased glycogenolysis, glucose uptake, glucose oxidation, and lactic acid production. Simultaneous deletion of glucose transporter (GLUT) 1 and GLUT3 (double knockout [DKO]) specifically in platelets completely abolished glucose uptake. In DKO platelets, mitochondrial oxidative metabolism of non-glycolytic substrates, such as glutamate, increased. Thrombosis and platelet activation were decreased through impairment at multiple activation nodes, including Ca2+ signaling, degranulation, and integrin activation. DKO mice developed thrombocytopenia, secondary to impaired pro-platelet formation from megakaryocytes, and increased platelet clearance resulting from cytosolic calcium overload and calpain activation. Systemic treatment with oligomycin, inhibiting mitochondrial metabolism, induced rapid clearance of platelets, with circulating counts dropping to zero in DKO mice, but not wild-type mice, demonstrating an essential role for energy metabolism in platelet viability. Thus, substrate metabolism is essential for platelet production, activation, and survival.
Collapse
Affiliation(s)
- Trevor P Fidler
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT 84112, USA; Program in Molecular Medicine, University of Utah, Salt Lake City, UT 84112, USA; Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Robert A Campbell
- Program in Molecular Medicine, University of Utah, Salt Lake City, UT 84112, USA; Department of Internal Medicine, University of Utah, Salt Lake City, UT 84112, USA
| | - Trevor Funari
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Nicholas Dunne
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Enrique Balderas Angeles
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Elizabeth A Middleton
- Program in Molecular Medicine, University of Utah, Salt Lake City, UT 84112, USA; Department of Internal Medicine, University of Utah, Salt Lake City, UT 84112, USA
| | - Dipayan Chaudhuri
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Andrew S Weyrich
- Program in Molecular Medicine, University of Utah, Salt Lake City, UT 84112, USA; Department of Internal Medicine, University of Utah, Salt Lake City, UT 84112, USA
| | - E Dale Abel
- Program in Molecular Medicine, University of Utah, Salt Lake City, UT 84112, USA; Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
89
|
Babur Ö, Ngo ATP, Rigg RA, Pang J, Rub ZT, Buchanan AE, Mitrugno A, David LL, McCarty OJT, Demir E, Aslan JE. Platelet procoagulant phenotype is modulated by a p38-MK2 axis that regulates RTN4/Nogo proximal to the endoplasmic reticulum: utility of pathway analysis. Am J Physiol Cell Physiol 2018; 314:C603-C615. [PMID: 29412690 PMCID: PMC6008067 DOI: 10.1152/ajpcell.00177.2017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 02/05/2018] [Accepted: 02/05/2018] [Indexed: 01/01/2023]
Abstract
Upon encountering physiological cues associated with damaged or inflamed endothelium, blood platelets set forth intracellular responses to ultimately support hemostatic plug formation and vascular repair. To gain insights into the molecular events underlying platelet function, we used a combination of interactome, pathway analysis, and other systems biology tools to analyze associations among proteins functionally modified by reversible phosphorylation upon platelet activation. While an interaction analysis mapped out a relative organization of intracellular mediators in platelet signaling, pathway analysis revealed directional signaling relations around protein kinase C (PKC) isoforms and mitogen-activated protein kinases (MAPKs) associated with platelet cytoskeletal dynamics, inflammatory responses, and hemostatic function. Pathway and causality analysis further suggested that platelets activate a specific p38-MK2 axis to phosphorylate RTN4 (reticulon-4, also known as Nogo), a Bcl-xl sequestration protein and critical regulator of endoplasmic reticulum (ER) physiology. In vitro, we find that platelets drive a p38-MK2-RTN4-Bcl-xl pathway associated with the regulation of the ER and platelet phosphatidylserine exposure. Together, our results support the use of pathway tools in the analysis of omics data sets as a means to help generate novel, mechanistic, and testable hypotheses for platelet studies while uncovering RTN4 as a putative regulator of platelet cell physiological responses.
Collapse
Affiliation(s)
- Özgün Babur
- Department of Molecular and Medical Genetics, Oregon Health & Science University , Portland, Oregon
- Computational Biology Program, Oregon Health & Science University , Portland, Oregon
| | - Anh T P Ngo
- Department of Biomedical Engineering, Oregon Health & Science University , Portland, Oregon
| | - Rachel A Rigg
- Department of Biomedical Engineering, Oregon Health & Science University , Portland, Oregon
| | - Jiaqing Pang
- Department of Biomedical Engineering, Oregon Health & Science University , Portland, Oregon
| | - Zhoe T Rub
- Department of Biomedical Engineering, Oregon Health & Science University , Portland, Oregon
| | - Ariana E Buchanan
- Knight Cardiovascular Institute, School of Medicine, Oregon Health & Science University , Portland, Oregon
| | - Annachiara Mitrugno
- Department of Biomedical Engineering, Oregon Health & Science University , Portland, Oregon
| | - Larry L David
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University , Portland, Oregon
| | - Owen J T McCarty
- Department of Biomedical Engineering, Oregon Health & Science University , Portland, Oregon
- Department of Cell, Developmental, & Cancer Biology, Oregon Health & Science University , Portland, Oregon
- Division of Hematology & Medical Oncology, Oregon Health & Science University , Portland, Oregon
| | - Emek Demir
- Department of Molecular and Medical Genetics, Oregon Health & Science University , Portland, Oregon
- Computational Biology Program, Oregon Health & Science University , Portland, Oregon
| | - Joseph E Aslan
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University , Portland, Oregon
- Knight Cardiovascular Institute, School of Medicine, Oregon Health & Science University , Portland, Oregon
| |
Collapse
|
90
|
14-3-3 proteins in platelet biology and glycoprotein Ib-IX signaling. Blood 2018; 131:2436-2448. [PMID: 29622550 DOI: 10.1182/blood-2017-09-742650] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 03/25/2018] [Indexed: 12/16/2022] Open
Abstract
Members of the 14-3-3 family of proteins function as adapters/modulators that recognize phosphoserine/phosphothreonine-based binding motifs in many intracellular proteins and play fundamental roles in signal transduction pathways of eukaryotic cells. In platelets, 14-3-3 plays a wide range of regulatory roles in phosphorylation-dependent signaling pathways, including G-protein signaling, cAMP signaling, agonist-induced phosphatidylserine exposure, and regulation of mitochondrial function. In particular, 14-3-3 interacts with several phosphoserine-dependent binding sites in the major platelet adhesion receptor, the glycoprotein Ib-IX complex (GPIb-IX), regulating its interaction with von Willebrand factor (VWF) and mediating VWF/GPIb-IX-dependent mechanosignal transduction, leading to platelet activation. The interaction of 14-3-3 with GPIb-IX also plays a critical role in enabling the platelet response to low concentrations of thrombin through cooperative signaling mediated by protease-activated receptors and GPIb-IX. The various functions of 14-3-3 in platelets suggest that it is a possible target for the treatment of thrombosis and inflammation.
Collapse
|
91
|
Varjú I, Farkas VJ, Kőhidai L, Szabó L, Farkas ÁZ, Polgár L, Chinopoulos C, Kolev K. Functional cyclophilin D moderates platelet adhesion, but enhances the lytic resistance of fibrin. Sci Rep 2018; 8:5366. [PMID: 29599453 PMCID: PMC5876378 DOI: 10.1038/s41598-018-23725-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 03/20/2018] [Indexed: 01/11/2023] Open
Abstract
In the course of thrombosis, platelets are exposed to a variety of activating stimuli classified as 'strong' (e.g. thrombin and collagen) or 'mild' (e.g. ADP). In response, activated platelets adhere to injured vasculature, aggregate, and stabilise the three-dimensional fibrin scaffold of the expanding thrombus. Since 'strong' stimuli also induce opening of the mitochondrial permeability transition pore (MPTP) in platelets, the MPTP-enhancer Cyclophilin D (CypD) has been suggested as a critical pharmacological target to influence thrombosis. However, it is poorly understood what role CypD plays in the platelet response to 'mild' stimuli which act independently of MPTP. Furthermore, it is unknown how CypD influences platelet-driven clot stabilisation against enzymatic breakdown (fibrinolysis). Here we show that treatment of human platelets with Cyclosporine A (a cyclophilin-inhibitor) boosts ADP-induced adhesion and aggregation, while genetic ablation of CypD in murine platelets enhances adhesion but not aggregation. We also report that platelets lacking CypD preserve their integrity in a fibrin environment, and lose their ability to render clots resistant against fibrinolysis. Our results indicate that CypD has opposing haemostatic roles depending on the stimulus and stage of platelet activation, warranting a careful design of any antithrombotic strategy targeting CypD.
Collapse
Affiliation(s)
- Imre Varjú
- Department of Medical Biochemistry, Semmelweis University, Budapest, 1094, Hungary
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, 02115, USA
- Department of Sociomedical Sciences, Mailman School of Public Health, Columbia University, New York, NY, 10032, USA
| | | | - László Kőhidai
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, 1089, Hungary
| | - László Szabó
- Department of Functional and Structural Materials, Institute of Materials and Environmental Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, 1117, Hungary
| | - Ádám Zoltán Farkas
- Department of Medical Biochemistry, Semmelweis University, Budapest, 1094, Hungary
| | - Lívia Polgár
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, 1089, Hungary
| | - Christos Chinopoulos
- Department of Medical Biochemistry, Semmelweis University, Budapest, 1094, Hungary
- MTA-SE Lendület Neurobiochemistry Research Group, Budapest, 1094, Hungary
| | - Krasimir Kolev
- Department of Medical Biochemistry, Semmelweis University, Budapest, 1094, Hungary.
| |
Collapse
|
92
|
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
|
93
|
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
|
94
|
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
|
95
|
Abstract
The human body generates 10-100 billion cells every day, and the same number of cells die to maintain homeostasis in our body. Cells infected by bacteria or viruses also die. The cell death that occurs under physiological conditions mainly proceeds by apoptosis, which is a noninflammatory, or silent, process, while pathogen infection induces necroptosis or pyroptosis, which activates the immune system and causes inflammation. Dead cells generated by apoptosis are quickly engulfed by macrophages for degradation. Caspases are a large family of cysteine proteases that act in cascades. A cascade that leads to caspase 3 activation mediates apoptosis and is responsible for killing cells, recruiting macrophages, and presenting an "eat me" signal(s). When apoptotic cells are not efficiently engulfed by macrophages, they undergo secondary necrosis and release intracellular materials that represent a damage-associated molecular pattern, which may lead to a systemic lupus-like autoimmune disease.
Collapse
Affiliation(s)
- Shigekazu Nagata
- Laboratory of Biochemistry and Immunology, World Premier International Research Center Initiative Immunology Frontier Research Center, Osaka University, Osaka 565-0871, Japan;
| |
Collapse
|
96
|
Apoptosis in megakaryocytes and platelets: the life and death of a lineage. Blood 2017; 131:605-610. [PMID: 29259001 DOI: 10.1182/blood-2017-11-742684] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 12/03/2017] [Indexed: 12/17/2022] Open
Abstract
Despite their profoundly different cellular composition, size, and function, megakaryocytes and platelets both depend on restraint of the intrinsic (or "mitochondrial") apoptosis pathway by BCL-2 family prosurvival proteins for their development and viability. Activation of the pathway contributes to the clearance of megakaryocytes following platelet shedding and constrains platelet lifespan in the circulation. Important questions remain as to how apoptosis is initiated in these cells at steady state and in response to pathophysiological insults.
Collapse
|
97
|
Nurden A. Platelets, inflammation and tissue regeneration. Thromb Haemost 2017; 105 Suppl 1:S13-33. [DOI: 10.1160/ths10-11-0720] [Citation(s) in RCA: 469] [Impact Index Per Article: 67.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2010] [Accepted: 02/04/2011] [Indexed: 12/20/2022]
Abstract
SummaryBlood platelets have long been recognised to bring about primary haemostasis with deficiencies in platelet production and function manifesting in bleeding while upregulated function favourises arterial thrombosis. Yet increasing evidence indicates that platelets fulfil a much wider role in health and disease. First, they store and release a wide range of biologically active substances including the panoply of growth factors, chemokines and cytokines released from α-granules. Membrane budding gives rise to microparticles (MPs), another active participant within the blood stream. Platelets are essential for the innate immune response and combat infection (viruses, bacteria, micro-organisms). They help maintain and modulate inflammation and are a major source of pro-inflammatory molecules (e.g. P-selectin, tissue factor, CD40L, metalloproteinases). As well as promoting coagulation, they are active in fibrinolysis; wound healing, angiogenesis and bone formation as well as in maternal tissue and foetal vascular remodelling. Activated platelets and MPs intervene in the propagation of major diseases. They are major players in atherosclerosis and related diseases, pathologies of the central nervous system (Alzheimers disease, multiple sclerosis), cancer and tumour growth. They participate in other tissue-related acquired pathologies such as skin diseases and allergy, rheumatoid arthritis, liver disease; while, paradoxically, autologous platelet-rich plasma and platelet releasate are being used as an aid to promote tissue repair and cellular growth. The above mentioned roles of platelets are now discussed.
Collapse
|
98
|
Abstract
Platelets are central to normal hemostasis and must be tightly controlled to prevent thrombosis. However, drug treatments that also affect platelets could lead to unwanted side effects on hemostasis or thrombosis. In this study, the effect of auranofin on platelets was tested. Auranofin, a gold-based thioredoxin reductase (TRXR) inhibitor, has been previously used in arthritis. Recently, auranofin and other inhibitors of the thioredoxin system have been proposed as novel anti-cancer therapies. TRXR is an important part of the antioxidant defenses in many cells that maintain intracellular proteins in their reduced state. TRXR activity in platelets could be completely inhibited by auranofin. Auranofin-treated platelets showed several features of cell death, including the inability to aggregate in response to thrombin, leakage of cytosolic lactate dehydrogenase, and surface exposure of procoagulant phosphatidylserine. Auranofin increased platelet reactive oxygen species production and intracellular calcium concentration. DTT, a sulfydyl reducing agent, and BAPTA-AM, which chelates intracellular calcium, prevented auranofin-induced phosphatidylserine exposure. These data suggest that TRXR is an important part of the platelet antioxidant defense. TRXR inhibition by auranofin triggers oxidative stress and disrupts intracellular calcium homeostasis, leading to platelet necrosis. The use of auranofin or other TRXR inhibitors could therefore lead to unwanted side effects.
Collapse
Affiliation(s)
- Matthew T Harper
- a Department of Pharmacology , University of Cambridge , CB2 1PD Cambridge , UK
| |
Collapse
|
99
|
Gao Q, Xiang Y, Chen Z, Zeng L, Ma X, Zhang Y. βγ-CAT, a non-lens betagamma-crystallin and trefoil factor complex, induces calcium-dependent platelet apoptosis. Thromb Haemost 2017; 105:846-54. [DOI: 10.1160/th10-10-0690] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2010] [Accepted: 01/29/2011] [Indexed: 11/05/2022]
Abstract
SummaryIn recent years, it has been reported that apoptosis may occur in platelets and play a role in the clearance of effete platelets. βγ-CAT, a newly identified non-lens βγ-crystallin and trefoil factor complex from frog Bombina maxima skin secretions, caused several in vivo toxic effects on mammals. Through determined haematological parameters of rabbits, it has been found that βγ-CAT significantly reduced the number of platelets in a time-dependent manner. Here, in order to explore the effect of βγ-CAT on platelets, washed platelets were incubated with various concentrations of βγ-CAT for 30 minutes. We found that βγ-CAT induced several apoptosis events in human platelets, including caspase-3 activation, phosphatidylserine (PS) exposure, depolarisation of mitochondrial inner transmembrane potential (ΔΨm), cytochrome c re-lease and strong expression of pro-apoptotic Bax and Bak proteins. However, βγ-CAT did not significantly induce platelet activation as detected by P-selectin surface expression, GPIIb/IIIa activation and platelet aggregation. In addition, we observed that βγ-CAT-induced PS exposure and ΔΨm depolarisation in platelets are Ca2+-dependent. Taken together, βγ-CAT can induce Ca2+-dependent platelet apoptosis but does not cause platelet activation.
Collapse
|
100
|
Alberio L, Ravanat C, Hechler B, Mangin PH, Lanza F, Gachet C. Delayed-onset of procoagulant signalling revealed by kinetic analysis of COAT platelet formation. Thromb Haemost 2017; 117:1101-1114. [DOI: 10.1160/th16-09-0711] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 03/19/2017] [Indexed: 11/05/2022]
Abstract
SummaryThe combined action of collagen and thrombin induces the formation of COAT platelets, which are characterised by a coat of procoagulant and adhesive molecules on their surface. Although recent work has started to highlight their clinical relevance, the exact mechanisms regulating the formation of procoagulant COAT platelets remain unclear. Therefore, we employed flow cytometry in order to visualise in real time surface and intracellular events following simultaneous platelet activation with convulxin and thrombin. After a rapid initial response pattern characterised by the homogenous activation of the fibrinogen receptor glycoprotein IIb/IIIa in all platelets, starting with a delay of about 2 minutes an increasing fraction transforms to procoagulant COAT platelets. Their surface is characterised by progressive loss of PAC-1 binding, expression of negative phospholipids and retention of α-granule von Willebrand factor. Intracellular events in procoagulant COAT platelets are a marked increase of free calcium into the low micromolar range, concomitantly with early depolarisation of the mitochondrial membrane and activation of caspase-3, while non-COAT platelets keep the intracellular free calcium in the nanomolar range and maintain an intact mitochondrial membrane. We show for the first time that the flow-cytometrically distinct fractions of COAT and non-COAT platelets differentially phosphorylate two signalling proteins, PKCα and p38MAPK, which may be involved in the regulation of the different calcium fluxes observed in COAT versus non-COAT platelets. This study demonstrates the utility of concomitant cellular and signalling evaluation using flow cytometry in order to further dissect the mechanisms underlying the dichotomous platelet response observed after collagen/thrombin stimulation.Supplementary Material to this article is available online at www.thrombosis-online.com.
Collapse
|