1
|
Mc Mahon A, Weiss L, Bennett K, Curley G, Ní Ainle F, Maguire P. Extracellular vesicles in disorders of hemostasis following traumatic brain injury. Front Neurol 2024; 15:1373266. [PMID: 38784907 PMCID: PMC11112090 DOI: 10.3389/fneur.2024.1373266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 04/29/2024] [Indexed: 05/25/2024] Open
Abstract
Traumatic brain injury (TBI) is a global health priority. In addition to being the leading cause of trauma related death, TBI can result in long-term disability and loss of health. Disorders of haemostasis are common despite the absence of some of the traditional risk factors for coagulopathy following trauma. Similar to trauma induced coagulopathy, this manifests with a biphasic response consisting of an early hypocoagulable phase and delayed hypercoagulable state. This coagulopathy is clinically significant and associated with increased rates of haemorrhagic expansion, disability and death. The pathophysiology of TBI-induced coagulopathy is complex but there is biologic plausibility and emerging evidence to suggest that extracellular vesicles (EVs) have a role to play. TBI and damage to the blood brain barrier result in release of brain-derived EVs that contain tissue factor and phosphatidylserine on their surface. This provides a platform on which coagulation can occur. Preclinical animal models have shown that an early rapid release of EVs results in overwhelming activation of coagulation resulting in a consumptive coagulopathy. This phenomenon can be attenuated with administration of substances to promote EV clearance and block their effects. Small clinical studies have demonstrated elevated levels of procoagulant EVs in patients with TBI correlating with clinical outcome. EVs represent a promising opportunity for use as minimally invasive biomarkers and potential therapeutic targets for TBI patients. However, additional research is necessary to bridge the gap between their potential and practical application in clinical settings.
Collapse
Affiliation(s)
- Aisling Mc Mahon
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
- Department of Critical Care Medicine, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Luisa Weiss
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
- SPHERE Research Group, Conway Institute, University College Dublin, Dublin, Ireland
| | - Kathleen Bennett
- Data Science Centre, School of Population Health, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Ger Curley
- Department of Anaesthesia and Critical Care Medicine, Beaumont Hospital, Dublin, Ireland
| | - Fionnuala Ní Ainle
- SPHERE Research Group, Conway Institute, University College Dublin, Dublin, Ireland
- Department of Haematology, Mater Misericordiae University Hospital and Rotunda Hospital, School of Medicine, University College Dublin, Dublin, Ireland
| | - Patricia Maguire
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
- SPHERE Research Group, Conway Institute, University College Dublin, Dublin, Ireland
- UCD Institute for Discovery, O'Brien Centre for Science, Dublin, Ireland
| |
Collapse
|
2
|
Zhou W, Zhao L, Mao Z, Wang Z, Zhang Z, Li M. Bidirectional Communication Between the Brain and Other Organs: The Role of Extracellular Vesicles. Cell Mol Neurobiol 2023; 43:2675-2696. [PMID: 37067749 PMCID: PMC10106324 DOI: 10.1007/s10571-023-01345-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 04/03/2023] [Indexed: 04/18/2023]
Abstract
A number of substances released by the brain under physiological and pathological conditions exert effects on other organs. In turn, substances produced primarily by organs such as bone marrow, adipose tissue, or the heart may have an impact on the metabolism and function and metabolism of the healthy and diseased brain. Despite a mounting amount of evidence supports such bidirectional communication between the brain and other organs, research on the function of molecular mediators carried by extracellular vesicles (EVs) is in the early stages. In addition to being able to target or reach practically any organ, EVs have the ability to cross the blood-brain barrier to transport a range of substances (lipids, peptides, proteins, and nucleic acids) to recipient cells, exerting biological effects. Here, we review the function of EVs in bidirectional communication between the brain and other organs. In a small number of cases, the role has been explicitly proven; yet, in most cases, it relies on indirect evidence from EVs in cell culture or animal models. There is a dearth of research currently available on the function of EVs-carrying mediators in the bidirectional communication between the brain and bone marrow, adipose tissue, liver, heart, lungs, and gut. Therefore, more studies are needed to determine how EVs facilitate communication between the brain and other organs.
Collapse
Affiliation(s)
- Wu Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, 17 Yongwai Street, Nanchang, 330006, Jiangxi, China
| | - Lihong Zhao
- Department of Radiotherapy, Jilin Cancer Hospital, 1018 Huguang Street, Changchun, 130012, Jilin, China
| | - Zelu Mao
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, 17 Yongwai Street, Nanchang, 330006, Jiangxi, China
| | - Zhihua Wang
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, 17 Yongwai Street, Nanchang, 330006, Jiangxi, China
| | - Zhixiong Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, 17 Yongwai Street, Nanchang, 330006, Jiangxi, China
| | - Meihua Li
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, 17 Yongwai Street, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
3
|
Gao Y, Li K, Li X, Li Q, Wang J, Zhang S, Zhang J. Exploration of cerebral vasospasm from the perspective of microparticles. Front Neurosci 2022; 16:1013437. [PMID: 36389239 PMCID: PMC9650082 DOI: 10.3389/fnins.2022.1013437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 10/14/2022] [Indexed: 02/28/2024] Open
Abstract
Cerebral vasospasm is a frequently encountered clinical problem, especially in patients with traumatic brain injury and subarachnoid hemorrhage. Continued cerebral vasospasm can cause cerebral ischemia, even infarction and delayed ischemic neurologic deficits. It significantly affects the course of the disease and the outcome of the patient. However, the underlying mechanism of cerebral vasospasm is still unclear. Recently, increasing studies focus on the pathogenic mechanism of microparticles. It has been found that microparticles have a non-negligible role in promoting vasospasm. This research aims to summarize the dynamics of microparticles in vivo and identify a causal role of microparticles in the occurrence and development of cerebral vasospasm. We found that these various microparticles showed dynamic characteristics in body fluids and directly or indirectly affect the cerebral vasospasm or prompt it. Due to the different materials carried by microparticles from different cells, there are also differences in the mechanisms that lead to abnormal vasomotor. We suggest that microparticle scavengers might be a promising therapeutic target against microparticles associated complications.
Collapse
Affiliation(s)
- Yalong Gao
- Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education and Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Kai Li
- Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education and Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaotian Li
- Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education and Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Qifeng Li
- Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education and Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Jiwei Wang
- Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education and Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin, China
| | - Shu Zhang
- Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education and Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Jianning Zhang
- Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education and Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
4
|
Neurovascular Unit-Derived Extracellular Vesicles: From Their Physiopathological Roles to Their Clinical Applications in Acute Brain Injuries. Biomedicines 2022; 10:biomedicines10092147. [PMID: 36140248 PMCID: PMC9495841 DOI: 10.3390/biomedicines10092147] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/26/2022] [Accepted: 08/28/2022] [Indexed: 11/16/2022] Open
Abstract
Extracellular vesicles (EVs) form a heterogeneous group of membrane-enclosed structures secreted by all cell types. EVs export encapsulated materials composed of proteins, lipids, and nucleic acids, making them a key mediator in cell–cell communication. In the context of the neurovascular unit (NVU), a tightly interacting multicellular brain complex, EVs play a role in intercellular communication and in maintaining NVU functionality. In addition, NVU-derived EVs can also impact peripheral tissues by crossing the blood–brain barrier (BBB) to reach the blood stream. As such, EVs have been shown to be involved in the physiopathology of numerous neurological diseases. The presence of NVU-released EVs in the systemic circulation offers an opportunity to discover new diagnostic and prognostic markers for those diseases. This review outlines the most recent studies reporting the role of NVU-derived EVs in physiological and pathological mechanisms of the NVU, focusing on neuroinflammation and neurodegenerative diseases. Then, the clinical application of EVs-containing molecules as biomarkers in acute brain injuries, such as stroke and traumatic brain injuries (TBI), is discussed.
Collapse
|
5
|
Sorah AB, Cunningham K, Wang H, Karvetski C, Ekaney M, Brintzenhoff R, Evans S. Effects of Guideline-Based Correction of Platelet Inhibition on Outcomes in Moderate to Severe Isolated Blunt Traumatic Brain Injury. Neurotrauma Rep 2022; 3:388-397. [PMID: 36204390 PMCID: PMC9531883 DOI: 10.1089/neur.2022.0003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Platelet dysfunction has been demonstrated after traumatic brain injury (TBI) regardless of the use of platelet inhibitors. The purpose of this study was to determine the efficacy of a platelet-mapping thromboelastography (PM-TEG) in predicting TBI patients who would benefit from platelet transfusion. We hypothesized that adenosine diphosphate (ADP) and arachadonic acid (AA) inhibition in patients with TBI is associated with increased mortality and can be corrected with platelet transfusion. This is a retrospective review of patients admitted to a level 1 trauma center from January 2016 through September 2017 with moderate to severe blunt TBI (msTBI), defined by an initial Glasgow Coma Scale (GCS) ≤12 with intracranial hemorrhage. Patients received PM-TEG. Those with platelet dysfunction (ADP or AA inhibition ≥60%) received one unit of platelets followed by repeat PM-TEG, until inhibition <60% or three units of platelets. Cohorts were defined as patients initially without (NPI) and with (PI) inhibition and subdivided into those whose inhibition corrected (PI-C) versus those whose did not correct (PI-NC). From 69 patients with isolated blunt TBI, 40 (58%) presented with NPI, 29 (42%) with PI. Of those with PI, 16 (55%) were with PI-C and 13 (45%) with PI-NC. Platelet inhibition in msTBI patients undergoing guideline-based transfusion is associated with age and GCS and an increase in mortality. Platelet inhibition seems to have a more adverse effect on patients >55 years of age or with GCS <8. Correction of platelet inhibition normalized mortality to that of NPI.
Collapse
Affiliation(s)
- Andrew B. Sorah
- F.H. Sammy Ross Trauma Center, Carolinas Medical Center, Charlotte, North Carolina, USA
| | - Kyle Cunningham
- F.H. Sammy Ross Trauma Center, Carolinas Medical Center, Charlotte, North Carolina, USA
| | | | - Colleen Karvetski
- Information and Analytic Services, Carolinas Medical Center, Charlotte, North Carolina, USA
| | - Michael Ekaney
- F.H. Sammy Ross Trauma Center, Carolinas Medical Center, Charlotte, North Carolina, USA
| | - Rita Brintzenhoff
- F.H. Sammy Ross Trauma Center, Carolinas Medical Center, Charlotte, North Carolina, USA
| | - Susan Evans
- F.H. Sammy Ross Trauma Center, Carolinas Medical Center, Charlotte, North Carolina, USA
| |
Collapse
|
6
|
Hubbard WB, Sim MMS, Saatman KE, Sullivan PG, Wood JP. Tissue factor release following traumatic brain injury drives thrombin generation. Res Pract Thromb Haemost 2022; 6:e12734. [PMID: 35702585 PMCID: PMC9175244 DOI: 10.1002/rth2.12734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 04/25/2022] [Accepted: 04/29/2022] [Indexed: 11/11/2022] Open
Abstract
Background Traumatic brain injury (TBI) results in neurovascular damage that initiates intrinsic mechanisms of hypercoagulation, which can contribute to the development of life-threatening complications, such as coagulopathy and delayed thrombosis. Clinical studies have hypothesized that tissue factor (TF) induces hypercoagulability after TBI; however, none have directly shown this relationship. Objectives In the current study, we took a stepwise approach to understand what factors are driving thrombin generation following experimental TBI. Methods We employed the contusion-producing controlled cortical impact (CCI) model and the diffuse closed head injury (CHI) model to investigate these mechanisms as a function of injury severity and modality. Whole blood was collected at 6 hours and 24 hours after injury, and platelet-poor plasma was used to measure thrombin generation and extracellular vesicle (EV) TF. Results We found that plasma thrombin generation, dependent on TF present in the plasma, was greater in CCI-injured animals compared to sham at both 6 hours (120.4 ± 36.9 vs 0.0 ± 0.0 nM*min endogenous thrombin potential) and 24 hours (131.0 ± 34.0 vs 32.1 ± 20.6 nM*min) after injury. This was accompanied by a significant increase in EV TF at 24 hours (328.6 ± 62.1 vs 167.7 ± 20.8 fM) after CCI. Further, EV TF is also increased at 6 hours (126.6 ± 17.1 vs 63.3 ± 14.4 fM) but not 24 hours following CHI. Conclusion TF-mediated thrombin generation is time-dependent after injury and TF increases resolve earlier following CHI as compared to CCI. Taken together, these data support a TF-mediated pathway of thrombin generation after TBI and pinpoint TF as a major player in TBI-induced coagulopathy.
Collapse
Affiliation(s)
- W. Brad Hubbard
- Lexington Veterans' Affairs Healthcare SystemLexingtonKentuckyUSA
- Department of PhysiologyUniversity of KentuckyLexingtonKentuckyUSA
- Spinal Cord and Brain Injury Research CenterUniversity of KentuckyLexingtonKentuckyUSA
| | - Martha M. S. Sim
- Department of Molecular and Cellular BiochemistryUniversity of KentuckyLexingtonKentuckyUSA
| | - Kathryn E. Saatman
- Department of PhysiologyUniversity of KentuckyLexingtonKentuckyUSA
- Spinal Cord and Brain Injury Research CenterUniversity of KentuckyLexingtonKentuckyUSA
| | - Patrick G. Sullivan
- Lexington Veterans' Affairs Healthcare SystemLexingtonKentuckyUSA
- Spinal Cord and Brain Injury Research CenterUniversity of KentuckyLexingtonKentuckyUSA
- Department of NeuroscienceUniversity of KentuckyLexingtonKentuckyUSA
| | - Jeremy P. Wood
- Department of Molecular and Cellular BiochemistryUniversity of KentuckyLexingtonKentuckyUSA
- Division of Cardiovascular MedicineThe Gill Heart and Vascular InstituteUniversity of KentuckyLexingtonKentuckyUSA
- Saha Cardiovascular Research CenterUniversity of KentuckyLexingtonKentuckyUSA
| |
Collapse
|
7
|
Abstract
Research into TBI biomarkers has accelerated rapidly in the past decade owing to the heterogeneous nature of TBI pathologies and management, which pose challenges to TBI evaluation, management, and prognosis. TBI biomarker proteins resulting from axonal, neuronal, or glial cell injuries are widely used and have been extensively studied. However, they might not pass the blood-brain barrier with sufficient amounts to be detected in peripheral blood specimens, and further might not be detectable in the cerebrospinal fluid owing to flow limitations triggered by the injury itself. Despite the advances in TBI research, there is an unmet clinical need to develop and identify novel TBI biomarkers that entirely correlate with TBI pathologies on the molecular level, including mild TBI, and further enable physicians to predict patient outcomes and allow researchers to test neuroprotective agents to limit the extents of injury. Although the extracellular vesicles have been identified and studied long ago, they have recently been revisited and repurposed as potential TBI biomarkers that overcome the many limitations of the traditional blood and CSF assays. Animal and human experiments demonstrated the accuracy of several types of exosomes and miRNAs in detecting mild, moderate, and severe TBI. In this paper, we provide a comprehensive review of the traditional TBI biomarkers that are helpful in clinical practice. Also, we highlight the emerging roles of exosomes and miRNA being the promising candidates under investigation of current research.
Collapse
|
8
|
Zhang CN, Li FJ, Zhao ZL, Zhang JN. The role of extracellular vesicles in traumatic brain injury-induced acute lung injury. Am J Physiol Lung Cell Mol Physiol 2021; 321:L885-L891. [PMID: 34549593 DOI: 10.1152/ajplung.00023.2021] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Acute lung injury (ALI), a common complication after traumatic brain injury (TBI), can evolve into acute respiratory distress syndrome (ARDS) and has a mortality rate of 30%-40%. Secondary ALI after TBI exhibits the following typical pathological features: infiltration of neutrophils into the alveolar and interstitial space, alveolar septal thickening, alveolar edema, and hemorrhage. Extracellular vesicles (EVs) were recently identified as key mediators in TBI-induced ALI. Due to their small size and lipid bilayer, they can pass through the disrupted blood-brain barrier (BBB) into the peripheral circulation and deliver their contents, such as genetic material and proteins, to target cells through processes such as fusion, receptor-mediated interactions, and uptake. Acting as messengers, EVs contribute to mediating brain-lung cross talk after TBI. In this review, we aim to summarize the mechanism of EVs in TBI-induced ALI, which may provide new ideas for clinical treatment.
Collapse
Affiliation(s)
- Chao-Nan Zhang
- Department of Neurosurgery, Tianjin Institute of Neurology, grid.412645.0Tianjin Medical University General Hospital, Tianjin, China
| | - Fan-Jian Li
- Department of Neurosurgery, Tianjin Institute of Neurology, grid.412645.0Tianjin Medical University General Hospital, Tianjin, China
| | - Zi-Long Zhao
- Department of Neurosurgery, Tianjin Institute of Neurology, grid.412645.0Tianjin Medical University General Hospital, Tianjin, China
| | - Jian-Ning Zhang
- Department of Neurosurgery, Tianjin Institute of Neurology, grid.412645.0Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
9
|
Sabet N, Soltani Z, Khaksari M. Multipotential and systemic effects of traumatic brain injury. J Neuroimmunol 2021; 357:577619. [PMID: 34058510 DOI: 10.1016/j.jneuroim.2021.577619] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 05/07/2021] [Accepted: 05/24/2021] [Indexed: 02/06/2023]
Abstract
Traumatic brain injury (TBI) is one of the leading causes of disability and mortality of people at all ages. Biochemical, cellular and physiological events that occur during primary injury lead to a delayed and long-term secondary damage that can last from hours to years. Secondary brain injury causes tissue damage in the central nervous system and a subsequent strong and rapid inflammatory response that may lead to persistent inflammation. However, this inflammatory response is not limited to the brain. Inflammatory mediators are transferred from damaged brain tissue to the bloodstream and produce a systemic inflammatory response in peripheral organs, including the cardiovascular, pulmonary, gastrointestinal, renal and endocrine systems. Complications of TBI are associated with its multiple and systemic effects that should be considered in the treatment of TBI patients. Therefore, in this review, an attempt was made to examine the systemic effects of TBI in detail. It is hoped that this review will identify the mechanisms of injury and complications of TBI, and open a window for promising treatment in TBI complications.
Collapse
Affiliation(s)
- Nazanin Sabet
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran; Department of Physiology and Pharmacology, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Zahra Soltani
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran; Department of Physiology and Pharmacology, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| | - Mohammad Khaksari
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
10
|
Savioli G, Ceresa IF, Caneva L, Gerosa S, Ricevuti G. Trauma-Induced Coagulopathy: Overview of an Emerging Medical Problem from Pathophysiology to Outcomes. MEDICINES (BASEL, SWITZERLAND) 2021; 8:16. [PMID: 33805197 PMCID: PMC8064317 DOI: 10.3390/medicines8040016] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 02/15/2021] [Accepted: 03/07/2021] [Indexed: 12/17/2022]
Abstract
Coagulopathy induced by major trauma is common, affecting approximately one-third of patients after trauma. It develops independently of iatrogenic, hypothermic, and dilutive causes (such as iatrogenic cause in case of fluid administration), which instead have a pejorative aspect on coagulopathy. Notwithstanding the continuous research conducted over the past decade on Trauma-Induced Coagulopathy (TIC), it remains a life-threatening condition with a significant impact on trauma mortality. We reviewed the current evidence regarding TIC diagnosis and pathophysiological mechanisms and summarized the different iterations of optimal TIC management strategies among which product resuscitation, potential drug administrations, and hemostatis-focused approaches. We have identified areas of ongoing investigation and controversy in TIC management.
Collapse
Affiliation(s)
- Gabriele Savioli
- Emergency Department, IRCCS Policlinico San Matteo, PhD University of Pavia, 27100 Pavia, Italy; (I.F.C.); (S.G.)
| | - Iride Francesca Ceresa
- Emergency Department, IRCCS Policlinico San Matteo, PhD University of Pavia, 27100 Pavia, Italy; (I.F.C.); (S.G.)
| | - Luca Caneva
- Anesthesia and Intensive Care, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy;
| | - Sebastiano Gerosa
- Emergency Department, IRCCS Policlinico San Matteo, PhD University of Pavia, 27100 Pavia, Italy; (I.F.C.); (S.G.)
| | - Giovanni Ricevuti
- Department of Drug Science, University of Pavia, 27100 Pavia, Italy;
- Saint Camillus International University of Health Sciences, 00152 Rome, Italy
| |
Collapse
|
11
|
Chen H, Jing Y, Xu Z, Yang D, Ju S, Guo Y, Tian H, Xue L. Upregulation of C Terminus of Hsc70-Interacting Protein Attenuates Apoptosis and Procoagulant Activity and Facilitates Brain Repair After Traumatic Brain Injury. Front Neurosci 2020; 14:925. [PMID: 33013306 PMCID: PMC7506102 DOI: 10.3389/fnins.2020.00925] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 08/11/2020] [Indexed: 12/16/2022] Open
Abstract
Traumatic brain injury (TBI) could highly induce coagulopathy through breaking the dynamic balance between coagulation and fibrinolysis systems, which may be a major contributor to the progressive secondary injury cascade that occurs after TBI. Phosphatase and tensin homolog deleted on chromosome 10 (PTEN) inhibition is reported to exert neuroprotection in TBI, making it a potential regulatory target involved in TBI-induced coagulation disorder. PTEN level is controlled in a major way by E3 ligase-mediated degradation through the ubiquitin-proteasome system. The C terminus of Hsc70-interacting protein (CHIP) has been shown to regulate proteasomal degradation and ubiquitination level of PTEN. In the present study, CHIP was overexpressed and knocked down in mouse brain microvascular endothelial cells (bEnd.3) and tissues during the early phase of TBI. In vitro cell proliferation, cell apoptosis, migration capacity, and invasion capacity were determined. The changes of procoagulant and apoptosis molecules after TBI were also detected as well as the micrangium density and blood-brain barrier permeability after in vivo TBI. In vitro results demonstrated that CHIP overexpression facilitated bEnd.3 cell proliferation, migration, and invasion and downregulated cell apoptosis and the expressions of procoagulant molecules through promoting PTEN ubiquitination in a simulated TBI model with stretch-induced injury treatment. In vivo experiments also demonstrated that CHIP overexpression suppressed post-TBI apoptosis and procoagulant protein expressions, as well as increased microvessel density, reduced hemorrhagic injury, and blood-brain barrier permeability. These findings suggested that the upregulation of CHIP may attenuate apoptosis and procoagulant activity, facilitate brain repair, and thus exerts neuroprotective effects in TBI.
Collapse
Affiliation(s)
- Hao Chen
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Yao Jing
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Zhiming Xu
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Dianxu Yang
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Shiming Ju
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Yan Guo
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Hengli Tian
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Lixia Xue
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| |
Collapse
|
12
|
Morris MC, John D, Singer KE, Moran R, McGlone E, Veile R, Goetzman HS, Makley AT, Caldwell CC, Goodman MD. Post-TBI splenectomy may exacerbate coagulopathy and platelet activation in a murine model. Thromb Res 2020; 193:211-217. [PMID: 32798961 DOI: 10.1016/j.thromres.2020.08.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/25/2020] [Accepted: 08/03/2020] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Traumatic brain injury (TBI) induces acute hypocoagulability, subacute hypercoagulability, and persistently elevated risk for thromboembolic events. Splenectomy is associated with increased mortality in patients with moderate or severe TBI. We hypothesized that the adverse effects of splenectomy in TBI patients may be secondary to the exacerbation of pathologic coagulation and platelet activation changes. METHODS An established murine weight-drop TBI model was utilized and a splenectomy was performed immediately following TBI. Sham as well as TBI and splenectomy alone mice were used as injury controls. Mice were sacrificed for blood draws at 1, 6, and 24 h, as well as 7 days post-TBI. Viscoelastic coagulation parameters were assessed by rotational thromboelastometry (ROTEM) and platelet activation was measured by expression of P-selectin via flow cytometry analysis of platelet rich plasma samples. RESULTS At 6 h following injury, TBI/splenectomy demonstrated hypocoagulability with prolonged clot formation time (CFT) compared to TBI alone. By 24 h following injury, TBI/splenectomy and splenectomy mice were hypercoagulable with a shorter CFT, a higher alpha angle, and increased MCF, despite a lower percentage of platelet contribution to clot compared to TBI alone. However, only the TBI/splenectomy continued to display this hypercoagulable state at 7 days. While TBI/splenectomy had greater P-selectin expression at 1-h post-injury, TBI alone had significantly greater P-selectin expression at 24 h post-injury compared to TBI/splenectomy. Interestingly, P-selectin expression remained elevated only in TBI/splenectomy at 7 days post-injury. CONCLUSION Splenectomy following TBI exacerbates changes in the post-injury coagulation state. The combination of TBI and splenectomy induces an acute hypocoagulable state that could contribute to an increase in intracranial bleeding. Subacutely, the addition of splenectomy to TBI exacerbates post-injury hypercoagulability and induces persistent platelet activation. These polytrauma effects on coagulation may contribute to the increased mortality observed in patients with combined brain and splenic injuries.
Collapse
Affiliation(s)
| | - Devin John
- Department of Surgery, University of Cincinnati, Cincinnati, OH, USA
| | - Kathleen E Singer
- Department of Surgery, University of Cincinnati, Cincinnati, OH, USA
| | - Ryan Moran
- Department of Surgery, University of Cincinnati, Cincinnati, OH, USA
| | - Emily McGlone
- Department of Surgery, University of Cincinnati, Cincinnati, OH, USA
| | - Rosalie Veile
- Department of Surgery, University of Cincinnati, Cincinnati, OH, USA
| | - Holly S Goetzman
- Department of Surgery, University of Cincinnati, Cincinnati, OH, USA; Division of Research, Shriners Hospital for Children, Cincinnati, OH, USA
| | - Amy T Makley
- Department of Surgery, University of Cincinnati, Cincinnati, OH, USA
| | - Charles C Caldwell
- Department of Surgery, University of Cincinnati, Cincinnati, OH, USA; Division of Research, Shriners Hospital for Children, Cincinnati, OH, USA
| | - Michael D Goodman
- Department of Surgery, University of Cincinnati, Cincinnati, OH, USA.
| |
Collapse
|
13
|
Relja B, Land WG. Damage-associated molecular patterns in trauma. Eur J Trauma Emerg Surg 2020; 46:751-775. [PMID: 31612270 PMCID: PMC7427761 DOI: 10.1007/s00068-019-01235-w] [Citation(s) in RCA: 119] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 09/27/2019] [Indexed: 12/13/2022]
Abstract
In 1994, the "danger model" argued that adaptive immune responses are driven rather by molecules released upon tissue damage than by the recognition of "strange" molecules. Thus, an alternative to the "self versus non-self recognition model" has been provided. The model, which suggests that the immune system discriminates dangerous from safe molecules, has established the basis for the future designation of damage-associated molecular patterns (DAMPs), a term that was coined by Walter G. Land, Seong, and Matzinger. The pathological importance of DAMPs is barely somewhere else evident as in the posttraumatic or post-surgical inflammation and regeneration. Since DAMPs have been identified to trigger specific immune responses and inflammation, which is not necessarily detrimental but also regenerative, it still remains difficult to describe their "friend or foe" role in the posttraumatic immunogenicity and healing process. DAMPs can be used as biomarkers to indicate and/or to monitor a disease or injury severity, but they also may serve as clinically applicable parameters for optimized indication of the timing for, i.e., secondary surgeries. While experimental studies allow the detection of these biomarkers on different levels including cellular, tissue, and circulatory milieu, this is not always easily transferable to the human situation. Thus, in this review, we focus on the recent literature dealing with the pathophysiological importance of DAMPs after traumatic injury. Since dysregulated inflammation in traumatized patients always implies disturbed resolution of inflammation, so-called model of suppressing/inhibiting inducible DAMPs (SAMPs) will be very briefly introduced. Thus, an update on this topic in the field of trauma will be provided.
Collapse
Affiliation(s)
- Borna Relja
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto von Guericke University Magdeburg, Magdeburg, Germany.
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Frankfurt, Goethe University Frankfurt am Main, 60590, Frankfurt, Germany.
| | - Walter Gottlieb Land
- Molecular ImmunoRheumatology, INSERM UMR_S1109, Laboratory of Excellence Transplantex, University of Strasbourg, Strasbourg, France
| |
Collapse
|
14
|
Yeates EO, Grigorian A, Schubl SD, Kuza CM, Joe V, Lekawa M, Borazjani B, Nahmias J. Chemoprophylaxis and Venous Thromboembolism in Traumatic Brain Injury at Different Trauma Centers. Am Surg 2020. [DOI: 10.1177/000313482008600433] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Patients with severe traumatic brain injury (TBI) are at an increased risk of venous thromboembolism (VTE). Because of concerns of worsening intracranial hemorrhage, clinicians are hesitant to start VTE chemoprophylaxis in this population. We hypothesized that ACS Level I trauma centers would be more aggressive with VTE chemoprophylaxis in adults with severe TBI than Level II centers. We also predicted that Level I centers would have a lower risk of VTE. We queried the Trauma Quality Improvement Program (2010–2016) database for patients with Abbreviated Injury Scale scores of 4 and 5 of the head and compared them based on treating the hospital trauma level. Of 204,895 patients with severe TBI, 143,818 (70.2%) were treated at Level I centers and 61,077 (29.8%) at Level II centers. The Level I cohort had a higher rate of VTE chemoprophylaxis use (43.2% vs 23.3%, P < 0.001) and a shorter median time to chemoprophylaxis (61.9 vs 85.9 hours, P < 0.001). Although Level I trauma centers started VTE chemoprophylaxis more often and earlier than Level II centers, there was no difference in the risk of VTE ( P = 0.414) after controlling for covariates. Future prospective studies are warranted to evaluate the timing, safety, and efficacy of early VTE chemoprophylaxis in severe TBI patients.
Collapse
Affiliation(s)
- Eric O. Yeates
- Department of Trauma, Burns and Surgical Critical Care, University of California, Irvine Medical Center, Orange, California; and
| | - Areg Grigorian
- Department of Trauma, Burns and Surgical Critical Care, University of California, Irvine Medical Center, Orange, California; and
| | - Sebastian D. Schubl
- Department of Trauma, Burns and Surgical Critical Care, University of California, Irvine Medical Center, Orange, California; and
| | - Catherine M. Kuza
- Department of Anesthesiology, University of Southern California Medical Center, Los Angeles, California
| | - Victor Joe
- Department of Trauma, Burns and Surgical Critical Care, University of California, Irvine Medical Center, Orange, California; and
| | - Michael Lekawa
- Department of Trauma, Burns and Surgical Critical Care, University of California, Irvine Medical Center, Orange, California; and
| | - Boris Borazjani
- Department of Trauma, Burns and Surgical Critical Care, University of California, Irvine Medical Center, Orange, California; and
| | - Jeffry Nahmias
- Department of Trauma, Burns and Surgical Critical Care, University of California, Irvine Medical Center, Orange, California; and
| |
Collapse
|
15
|
Microvesicles generated following traumatic brain injury induce platelet dysfunction via adenosine diphosphate receptor. J Trauma Acute Care Surg 2020; 86:592-600. [PMID: 30614923 DOI: 10.1097/ta.0000000000002171] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Traumatic brain injury (TBI) can result in an acute coagulopathy including platelet dysfunction that can contribute to ongoing intracranial hemorrhage. Previous studies have shown adenosine diphosphate (ADP)-induced platelet aggregation to be reduced after TBI. In addition, circulating microvesicles (MVs) are increased following TBI and have been shown to play a role in post-TBI coagulopathy and platelet function. We hypothesized that post-TBI MVs would affect platelet aggregation in a murine head injury model. METHODS Moderate TBI was performed using a weight-drop method in male C57BL6 mice. Whole blood, plasma, MVs, and MV-poor plasma were isolated from blood collected 10 minutes following TBI and were mixed separately with whole blood from uninjured mice. Platelet aggregation was measured with Multiplate impedance platelet aggregometry in response to ADP. The ADP P2Y12 receptor inhibitor, R-138727, was incubated with plasma and MVs from TBI mice, and platelet inhibition was again measured. RESULTS Whole blood taken from 10-minute post-TBI mice demonstrated diminished ADP-induced platelet aggregation compared with sham mice. When mixed with normal donor blood, post-TBI plasma and MVs induced diminished ADP-induced platelet aggregation compared with sham plasma and sham MVs. By contrast, the addition of post-TBI MV-poor plasma to normal blood did not change ADP-induced platelet aggregation. The observed dysfunction in post-TBI ADP platelet aggregation was prevented by the pretreatment of post-TBI plasma with R-138727. Treatment of post-TBI MVs with R-138727 resulted in similar findings of improved ADP-induced platelet aggregation compared with nontreated post-TBI MVs. CONCLUSION Adenosine diphosphate-induced platelet aggregation is inhibited acutely following TBI in a murine model. This platelet inhibition is reproduced in normal blood by the introduction of post-TBI plasma and MVs. Furthermore, observed platelet dysfunction is prevented when post-TBI plasma and MVs are treated with an inhibitor of the P2Y12 ADP receptor. Clinically observed post-TBI platelet dysfunction may therefore be partially explained by the presence of the ADP P2Y12 receptor within post-TBI MVs. LEVEL OF EVIDENCE Level III.
Collapse
|
16
|
Neuroprotective functions of calycosin against intracerebral hemorrhage-induced oxidative stress and neuroinflammation. Future Med Chem 2020; 12:583-592. [PMID: 32116027 DOI: 10.4155/fmc-2019-0311] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Aim: To investigate whether calycosin affects the brain damages induced by intracerebral hemorrhage (ICH). Methods: ICH mouse model was established by injection of collagenase type VII. Results: 50 mg/kg calycosin showed significant inhibitory effects on ICH-induced brain impairment evaluated by modified neurologic severity scores and water content. In addition, the lesion volumes, blood accumulation and hemispheric enlargement were all dramatically reduced by calycosin treatment compared with those of vehicles. It was observed that calycosin repressed oxidative stress by enhancing Nrf2 anti-oxidative pathway and suppressed inflammation by blocking NACHT, NALP3 inflammasome and NF-κB pathway activation. Conclusion: Calycosin could protect the brain against the damages induced by ICH via inhibiting oxidative damages and inflammation.
Collapse
|
17
|
Chen Z, Chopp M, Zacharek A, Li W, Venkat P, Wang F, Landschoot-Ward J, Chen J. Brain-Derived Microparticles (BDMPs) Contribute to Neuroinflammation and Lactadherin Reduces BDMP Induced Neuroinflammation and Improves Outcome After Stroke. Front Immunol 2019; 10:2747. [PMID: 31993045 PMCID: PMC6968774 DOI: 10.3389/fimmu.2019.02747] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 11/08/2019] [Indexed: 12/28/2022] Open
Abstract
Microparticles (MPs, ~size between 0.1 and 1 mm) are lipid encased containers derived from intact cells which contain antigen from the parent cells. MPs are involved in intercellular communication and regulate inflammation. Stroke increases secretion of brain derived MP (BDMP) which activate macrophages/microglia and induce neuroinflammation. Lactadherin (Milk fat globule–EGF factor-8) binds to anionic phospholipids and extracellular matrices, promotes apoptotic cell clearance and limits pathogenic antigen cross presentation. In this study, we investigate whether BDMP affects stroke-induced neuroinflammation and whether Lactadherin treatment reduces stroke initiated BDMP-induced neuroinflammation, thereby improving functional outcome after stroke. Middle aged (8–9 months old) male C57BL/6J mice were subjected to distal middle cerebral artery occlusion (dMCAo) stroke, and BDMPs were extracted from ischemic brain 24 h after dMCAo by ultracentrifugation. Adult male C57BL/6J mice were subjected to dMCAo and treated via tail vein injection at 3 h after stroke with: (A) +PBS (n = 5/group); (B) +BDMPs (1.5 × 108, n = 6/group); (C) +Lactadherin (400 μg/kg, n = 5/group); (D) +BDMP+Lactadherin (n = 6/group). A battery of neurological function tests were performed and mice sacrificed for immunostaining at 14 days after stroke. Blood plasma was used for Western blot assay. Our data indicate: (1) treatment of Stroke with BDMP significantly increases lesion volume, neurological deficits, blood brain barrier (BBB) leakage, microglial activation, inflammatory cell infiltration (CD45, microglia/macrophages, and neutrophils) into brain, inflammatory factor (TNFα, IL6, and IL1β) expression in brain, increases axon/white matter (WM) damage identified by decreased axon and myelin density, and increases inflammatory factor expression in the plasma when compared to PBS treated stroke mice; (2) when compared to PBS and BDMP treated stroke mice, Lactadherin and BDMP+Lactadherin treatment significantly improves neurological outcome, and decreases lesion volume, BBB leakage, axon/WM injury, inflammatory cell infiltration and inflammatory factor expression in the ischemic brain, respectively. Lactadherin treatment significantly increases anti-inflammatory factor (IL10) expression in ischemic brain and decreases IL1β expression in plasma compared to PBS and BDMP treated stroke mice, respectively. BDMP increases neuroinflammation and aggravates ischemic brain damage after stroke. Thus, Lactadherin exerts anti-inflammatory effects and improves the clearance of MPs to reduce stroke and BDMP induced neurological deficits.
Collapse
Affiliation(s)
- Zhili Chen
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States.,Department of Physics, Oakland University, Rochester, MI, United States
| | - Alex Zacharek
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Wei Li
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Poornima Venkat
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Fenjie Wang
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | | | - Jieli Chen
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| |
Collapse
|
18
|
Yates AG, Anthony DC, Ruitenberg MJ, Couch Y. Systemic Immune Response to Traumatic CNS Injuries-Are Extracellular Vesicles the Missing Link? Front Immunol 2019; 10:2723. [PMID: 31824504 PMCID: PMC6879545 DOI: 10.3389/fimmu.2019.02723] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 11/06/2019] [Indexed: 12/16/2022] Open
Abstract
Inflammation following traumatic injury to the central nervous system (CNS) persists long after the primary insult and is known to exacerbate cell death and worsen functional outcomes. Therapeutic interventions targeting this inflammation have been unsuccessful, which has been attributed to poor bioavailability owing to the presence of blood-CNS barrier. Recent studies have shown that the magnitude of the CNS inflammatory response is dependent on systemic inflammatory events. The acute phase response (APR) to CNS injury presents an alternative strategy to modulating the secondary phase of injury. However, the communication pathways between the CNS and the periphery remain poorly understood. Extracellular vesicles (EVs) are membrane bound nanoparticles that are regulators of intercellular communication. They are shed from cells of the CNS including microglia, astrocytes, neurons and endothelial cells, and are able to cross the blood-CNS barrier, thus providing an attractive candidate for initiating the APR after acute CNS injury. The purpose of this review is to summarize the current evidence that EVs play a critical role in the APR following CNS injuries.
Collapse
Affiliation(s)
- Abi G Yates
- Department of Pharmacology, Medical Sciences Division, University of Oxford, Oxford, United Kingdom.,School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Daniel C Anthony
- Department of Pharmacology, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Marc J Ruitenberg
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Yvonne Couch
- Acute Stroke Programme, RDM-Investigative Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
19
|
Robicsek SA, Bhattacharya A, Rabai F, Shukla K, Doré S. Blood-Related Toxicity after Traumatic Brain Injury: Potential Targets for Neuroprotection. Mol Neurobiol 2019; 57:159-178. [PMID: 31617072 DOI: 10.1007/s12035-019-01766-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 08/29/2019] [Indexed: 02/07/2023]
Abstract
Emergency visits, hospitalizations, and deaths due to traumatic brain injury (TBI) have increased significantly over the past few decades. While the primary early brain trauma is highly deleterious to the brain, the secondary injury post-TBI is postulated to significantly impact mortality. The presence of blood, particularly hemoglobin, and its breakdown products and key binding proteins and receptors modulating their clearance may contribute significantly to toxicity. Heme, hemin, and iron, for example, cause membrane lipid peroxidation, generate reactive oxygen species, and sensitize cells to noxious stimuli resulting in edema, cell death, and increased morbidity and mortality. A wide range of other mechanisms such as the immune system play pivotal roles in mediating secondary injury. Effective scavenging of all of these pro-oxidant and pro-inflammatory metabolites as well as controlling maladaptive immune responses is essential for limiting toxicity and secondary injury. Hemoglobin metabolism is mediated by key molecules such as haptoglobin, heme oxygenase, hemopexin, and ferritin. Genetic variability and dysfunction affecting these pathways (e.g., haptoglobin and heme oxygenase expression) have been implicated in the difference in susceptibility of individual patients to toxicity and may be target pathways for potential therapeutic interventions in TBI. Ongoing collaborative efforts are required to decipher the complexities of blood-related toxicity in TBI with an overarching goal of providing effective treatment options to all patients with TBI.
Collapse
Affiliation(s)
- Steven A Robicsek
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, College of Medicine, University of Florida, 1275 Center Drive, Biomed Sci J493, Gainesville, FL, 32610, USA. .,Departments of Neurosurgery, Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA.
| | - Ayon Bhattacharya
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, College of Medicine, University of Florida, 1275 Center Drive, Biomed Sci J493, Gainesville, FL, 32610, USA.,Department of Pharmacology, KPC Medical College, West Bengal University of Health Sciences, Kolkata, West Bengal, India
| | - Ferenc Rabai
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, College of Medicine, University of Florida, 1275 Center Drive, Biomed Sci J493, Gainesville, FL, 32610, USA
| | - Krunal Shukla
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, College of Medicine, University of Florida, 1275 Center Drive, Biomed Sci J493, Gainesville, FL, 32610, USA
| | - Sylvain Doré
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, College of Medicine, University of Florida, 1275 Center Drive, Biomed Sci J493, Gainesville, FL, 32610, USA. .,Departments of Neurology, Psychiatry, Pharmaceutics and Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
20
|
Karttunen J, Heiskanen M, Lipponen A, Poulsen D, Pitkänen A. Extracellular Vesicles as Diagnostics and Therapeutics for Structural Epilepsies. Int J Mol Sci 2019; 20:E1259. [PMID: 30871144 PMCID: PMC6470789 DOI: 10.3390/ijms20061259] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 03/08/2019] [Accepted: 03/08/2019] [Indexed: 12/16/2022] Open
Abstract
Extracellular vesicles (EVs) are small vesicles involved in intercellular communication. Data is emerging that EVs and their cargo have potential as diagnostic biomarkers and treatments for brain diseases, including traumatic brain injury and epilepsy. Here, we summarize the current knowledge regarding changes in EV numbers and cargo in status epilepticus (SE) and traumatic brain injury (TBI), which are clinically significant etiologies for acquired epileptogenesis in animals and humans. We also review encouraging data, which suggests that EVs secreted by stem cells may serve as recovery-enhancing treatments for SE and TBI. Using Gene Set Enrichment Analysis, we show that brain EV-related transcripts are positively enriched in rodent models of epileptogenesis and epilepsy, and altered in response to anti-seizure drugs. These data suggest that EVs show promise as biomarkers, treatments and drug targets for epilepsy. In parallel to gathering conceptual knowledge, analytics platforms for the isolation and analysis of EV contents need to be further developed.
Collapse
Affiliation(s)
- Jenni Karttunen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland.
| | - Mette Heiskanen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland.
| | - Anssi Lipponen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland.
| | - David Poulsen
- University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Clinical and Translational Research Center (CTRC), Department of Neurosurgery, Buffalo, NY 14203, USA.
| | - Asla Pitkänen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland.
| |
Collapse
|
21
|
Dukhinova M, Kuznetsova I, Kopeikina E, Veniaminova E, Yung AWY, Veremeyko T, Levchuk K, Barteneva NS, Wing-Ho KK, Yung WH, Liu JYH, Rudd J, Yau SSY, Anthony DC, Strekalova T, Ponomarev ED. Platelets mediate protective neuroinflammation and promote neuronal plasticity at the site of neuronal injury. Brain Behav Immun 2018; 74:7-27. [PMID: 30217533 DOI: 10.1016/j.bbi.2018.09.009] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 09/10/2018] [Accepted: 09/10/2018] [Indexed: 01/05/2023] Open
Abstract
It is generally accepted that inflammation within the CNS contributes to neurodegeneration after traumatic brain injury (TBI), but it is not clear how inflammation is initiated in the absence of infection and whether this neuroinflammation is predominantly beneficial or detrimental. We have previously found that brain-enriched glycosphingolipids within neuronal lipid rafts (NLR) induced platelet degranulation and secretion of neurotransmitters and pro-inflammatory factors. In the present study, we compared TBI-induced inflammation and neurodegeneration in wild-type vs. St3gal5 deficient (ST3-/-) mice that lack major CNS-specific glycosphingolipids. After TBI, microglial activation and CNS macrophage infiltration were substantially reduced in ST3-/- animals. However, ST3-/- mice had a larger area of CNS damage with marked neuronal/axonal loss. The interaction of platelets with NLR stimulated neurite growth, increased the number of PSD95-positive dendritic spines, and intensified neuronal activity. Adoptive transfer and blocking experiments provide further that platelet-derived serotonin and platelet activating factor plays a key role in the regulation of sterile neuroinflammation, hemorrhage and neuronal plasticity after TBI.
Collapse
Affiliation(s)
- Marina Dukhinova
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin N.T., Hong Kong
| | - Inna Kuznetsova
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin N.T., Hong Kong
| | - Ekaterina Kopeikina
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin N.T., Hong Kong
| | - Ekaterina Veniaminova
- Department of Neuroscience, Maastricht University, Universiteitssingel 40, NL 6229ER, Maastricht, Netherlands; Institute of General Pathology and Pathophysiology, Baltiiskaya str, 8, Moscow, 125315, Russia; Sechenov First Moscow State Medical University, Institute of Molecular Medicine, Laboratory of Psychiatric Neurobiology, Trubetskaya Street 8-2, 119991, Moscow, Russia
| | - Amanda W Y Yung
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin N.T., Hong Kong
| | - Tatyana Veremeyko
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin N.T., Hong Kong
| | - Kseniia Levchuk
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin N.T., Hong Kong
| | - Natasha S Barteneva
- Program in Cellular and Molecular Medicine, Children's Hospital Boston and Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Kenny Kam Wing-Ho
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin N.T., Hong Kong
| | - Wing-Ho Yung
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin N.T., Hong Kong
| | - Julia Y H Liu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin N.T., Hong Kong
| | - John Rudd
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin N.T., Hong Kong; Brain and Mind Institute, The Chinese University of Hong Kong, Shatin NT, Hong Kong
| | - Sonata S Y Yau
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - Daniel C Anthony
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | - Tatyana Strekalova
- Department of Neuroscience, Maastricht University, Universiteitssingel 40, NL 6229ER, Maastricht, Netherlands; Institute of General Pathology and Pathophysiology, Baltiiskaya str, 8, Moscow, 125315, Russia; Sechenov First Moscow State Medical University, Institute of Molecular Medicine, Laboratory of Psychiatric Neurobiology, Trubetskaya Street 8-2, 119991, Moscow, Russia
| | - Eugene D Ponomarev
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin N.T., Hong Kong; Kunming Institute of Zoology and Chinese University of Hong Kong Joint Laboratory of Bioresources and Molecular Research of Common Diseases, Kunmin-Hong Kong, China.
| |
Collapse
|
22
|
Dore E, Boilard E. Roles of secreted phospholipase A 2 group IIA in inflammation and host defense. Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1864:789-802. [PMID: 30905346 DOI: 10.1016/j.bbalip.2018.08.017] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 08/29/2018] [Accepted: 08/30/2018] [Indexed: 01/08/2023]
Abstract
Among all members of the secreted phospholipase A2 (sPLA2) family, group IIA sPLA2 (sPLA2-IIA) is possibly the most studied enzyme. Since its discovery, many names have been associated with sPLA2-IIA, such as "non-pancreatic", "synovial", "platelet-type", "inflammatory", and "bactericidal" sPLA2. Whereas the different designations indicate comprehensive functions or sources proposed for this enzyme, the identification of the precise roles of sPLA2-IIA has remained a challenge. This can be attributed to: the expression of the enzyme by various cells of different lineages, its limited activity towards the membranes of immune cells despite its expression following common inflammatory stimuli, its ability to interact with certain proteins independently of its catalytic activity, and its absence from multiple commonly used mouse models. Nevertheless, elevated levels of the enzyme during inflammatory processes and associated consistent release of arachidonic acid from the membrane of extracellular vesicles suggest that sPLA2-IIA may contribute to inflammation by using endogenous substrates in the extracellular milieu. Moreover, the remarkable potency of sPLA2-IIA towards bacterial membranes and its induced expression during the course of infections point to a role for this enzyme in the defense of the host against invading pathogens. In this review, we present current knowledge related to mammalian sPLA2-IIA and its roles in sterile inflammation and host defense.
Collapse
Affiliation(s)
- Etienne Dore
- Centre de Recherche du CHU de Québec, Université Laval, Department of Infectious Diseases and Immunity, Québec City, QC, Canada
| | - Eric Boilard
- Centre de Recherche du CHU de Québec, Université Laval, Department of Infectious Diseases and Immunity, Québec City, QC, Canada; Canadian National Transplantation Research Program, Edmonton, AB, Canada.
| |
Collapse
|
23
|
Kuharić J, Grabušić K, Tokmadžić VS, Štifter S, Tulić K, Shevchuk O, Lučin P, Šustić A. Severe Traumatic Brain Injury Induces Early Changes in the Physical Properties and Protein Composition of Intracranial Extracellular Vesicles. J Neurotrauma 2018; 36:190-200. [PMID: 29690821 DOI: 10.1089/neu.2017.5515] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Extracellular vesicles (EVs) are membranous nanostructures that can indicate undergoing processes in organs and thus help in diagnostics and prognostics. They are secreted by all cells, contained in body fluids, and able to transfer proteins, lipids and nucleic acids to distant cells. Intracranial EVs were shown to change their composition after severe traumatic brain injury (TBI) and therefore to have biomarker potential to evaluate brain events. Properties of intracranial EVs early after TBI, however, have not been characterized. Here, we assessed cerebrospinal fluid (CSF) up to seven days after isolated severe TBI for physical properties of EVs and their proteins associated with neuroregeneration. These findings were compared with healthy controls and correlated to patient outcome. The study included 17 patients with TBI and 18 healthy controls. EVs in TBI-CSF were visualized by electron microscopy and confirmed by immunoblotting for membrane associated Flotillin-1 and Flotillin-2. Using nanoparticle tracking analysis, we detected the highest range in EV concentration at day 1 after injury and significantly increased EV size at days 4-7. CSF concentrations of neuroregeneration associated proteins Flotillin-1, ADP-ribosylation Factor 6 (Arf6), and Ras-related protein Rab7a (Rab7a) were monitored by enzyme-linked immunosorbent assays. Flotillin-1 was detected solely in TBI-CSF in about one third of tested patients. Unfavorable outcomes included decreasing Arf6 concentrations and a delayed Rab7a concentration increase in CSF. CSF concentrations of Arf6 and Rab7a were negatively correlated. Our data suggest that the brain response within several days after severe TBI includes shedding of EVs associated with neuroplasticity. Extended studies with a larger number of participants and CSF collected at shorter intervals are necessary to further evaluate neuroregeneration biomarker potential of Rab7a, Arf6, and Flotillin-1.
Collapse
Affiliation(s)
- Janja Kuharić
- 1 Department of Anaesthesia, Resuscitation and Intensive Care Medicine, Faculty of Medicine, University of Rijeka, Rijeka, Croatia.,2 Department of Anaesthesia and Intensive Care Medicine, Clinical Hospital Center Rijeka, Rijeka, Croatia
| | - Kristina Grabušić
- 3 Department of Biotechnology, University of Rijeka, Rijeka, Croatia
| | - Vlatka Sotošek Tokmadžić
- 1 Department of Anaesthesia, Resuscitation and Intensive Care Medicine, Faculty of Medicine, University of Rijeka, Rijeka, Croatia.,2 Department of Anaesthesia and Intensive Care Medicine, Clinical Hospital Center Rijeka, Rijeka, Croatia.,4 Department of Clinical Medical Sciences II, Faculty of Health Studies, University of Rijeka, Rijeka, Croatia
| | - Sanja Štifter
- 5 Department of General Pathology and Pathological Anatomy, Faculty of Medicine, University of Rijeka, Rijeka, Croatia.,6 Department of Pathology, Clinical Hospital Center Rijeka, Rijeka, Croatia
| | - Ksenija Tulić
- 7 Department of Physiology and Immunology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Olga Shevchuk
- 8 Leibniz Institut für Analytische Wissenschaften, ISAS Campus, Dortmund, Germany
| | - Pero Lučin
- 7 Department of Physiology and Immunology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Alan Šustić
- 1 Department of Anaesthesia, Resuscitation and Intensive Care Medicine, Faculty of Medicine, University of Rijeka, Rijeka, Croatia.,2 Department of Anaesthesia and Intensive Care Medicine, Clinical Hospital Center Rijeka, Rijeka, Croatia.,4 Department of Clinical Medical Sciences II, Faculty of Health Studies, University of Rijeka, Rijeka, Croatia
| |
Collapse
|
24
|
Mondello S, Thelin EP, Shaw G, Salzet M, Visalli C, Cizkova D, Kobeissy F, Buki A. Extracellular vesicles: pathogenetic, diagnostic and therapeutic value in traumatic brain injury. Expert Rev Proteomics 2018; 15:451-461. [PMID: 29671356 DOI: 10.1080/14789450.2018.1464914] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Traumatic brain injury (TBI) is a leading cause of death and disability worldwide. Accurate classification according to injury-specific and patient-specific characteristics is critical to help informed clinical decision-making and to the pursuit of precision medicine in TBI. Reliable biomarker signatures for improved TBI diagnostics are required but still an unmet need. Areas covered: Extracellular vesicles (EVs) represent a new class of biomarker candidates in TBI. These nano-sized vesicles have key roles in cell signaling profoundly impacting pathogenic pathways, progression and long-term sequelae of TBI. As such EVs might provide novel neurobiological insights, enhance our understanding of the molecular mechanisms underlying TBI pathophysiology and recovery, and serve as biomarker signatures and therapeutic targets and delivery systems. Expert commentary: EVs are fast gaining momentum in TBI research, paving the way for new transformative diagnostic and treatment approaches. Their potential to sort out TBI variability and active involvement in the mechanisms underpinning different clinical phenotypes point out unique opportunities for improved classification, risk-stratification ad intervention, harboring promise of predictive, personalized, and even preemptive therapeutic strategies. Although a great deal of progress has been made, substantial efforts are still required to ensure the needed rigorous validation and reproducibility for clinical implementation of EVs.
Collapse
Affiliation(s)
- Stefania Mondello
- a Oasi Research Institute-IRCCS , Troina , Italy.,b Department of Biomedical and Dental Sciences and Morphofunctional Imaging , University of Messina , Messina , Italy
| | - Eric P Thelin
- c Department of Clinical Neuroscience , Karolinska Institutet , Stockholm , Sweden.,d Department of Clinical Neurosciences, Division of Neurosurgery , University of Cambridge, Cambridge Biomedical Campus , Cambridge , United Kingdom
| | - Gerry Shaw
- e EnCor Biotechnology Inc ., Gainesville , FL , USA
| | - Michel Salzet
- f Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), Université de Lille , Lille , France
| | - Carmela Visalli
- b Department of Biomedical and Dental Sciences and Morphofunctional Imaging , University of Messina , Messina , Italy
| | - Dasa Cizkova
- g Neuroimmunology , Slovak Academy of Sciences , Bratislava , Slovakia
| | - Firas Kobeissy
- h Department of Psychiatry and Neuroscience , McKnight Brain Institute, University of Florida , Gainesville , FL , USA.,i Department of Biochemistry and Molecular Genetics , American University of Beirut , Beirut , Lebanon
| | - Andras Buki
- j Department of Neurosurgery , Pecs University , Pecs , Hungary
| |
Collapse
|
25
|
Boilard E. Extracellular vesicles and their content in bioactive lipid mediators: more than a sack of microRNA. J Lipid Res 2018; 59:2037-2046. [PMID: 29678959 DOI: 10.1194/jlr.r084640] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 03/26/2018] [Indexed: 01/08/2023] Open
Abstract
Extracellular vesicles (EVs), such as exosomes and microvesicles, are small membrane-bound vesicles released by cells under various conditions. In a multitude of physiological and pathological conditions, EVs contribute to intercellular communication by facilitating exchange of material between cells. Rapidly growing interest is aimed at better understanding EV function and their use as biomarkers. The vast EV cargo includes cytokines, growth factors, organelles, nucleic acids (messenger and micro RNA), and transcription factors. A large proportion of research dedicated to EVs is focused on their microRNA cargo; however, much less is known about other EV constituents, in particular, eicosanoids. These potent bioactive lipid mediators, derived from arachidonic acid, are shuttled in EVs along with the enzymes in charge of their synthesis. In the extracellular milieu, EVs also interact with secreted phospholipases to generate eicosanoids, which then regulate the transfer of cargo into a cellular recipient. Eicosanoids are useful as biomarkers and contribute to a variety of biological functions, including modulation of distal immune responses. Here, we review the reported roles of eicosanoids conveyed by EVs and describe their potential as biomarkers.
Collapse
Affiliation(s)
- Eric Boilard
- Centre de Recherche du CHU de Québec - Université Laval, Department of Infectious Diseases and Immunity, Quebec City, QC, Canada, and Canadian National Transplantation Research Program, Edmonton, AB, Canada
| |
Collapse
|
26
|
Coagulopathy induced by traumatic brain injury: systemic manifestation of a localized injury. Blood 2018; 131:2001-2006. [PMID: 29507078 DOI: 10.1182/blood-2017-11-784108] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 02/22/2018] [Indexed: 12/15/2022] Open
Abstract
Traumatic brain injury (TBI)-induced coagulopathy is a common and well-recognized risk for poor clinical outcomes, but its pathogenesis remains poorly understood, and treatment options are limited and ineffective. We discuss the recent progress and knowledge gaps in understanding this lethal complication of TBI. We focus on (1) the disruption of the brain-blood barrier to disseminate brain injury systemically by releasing brain-derived molecules into the circulation and (2) TBI-induced hypercoagulable and hyperfibrinolytic states that result in persistent and delayed intracranial hemorrhage and systemic bleeding.
Collapse
|
27
|
Kuravi SJ, Yates CM, Foster M, Harrison P, Hazeldine J, Hampson P, Watson C, Belli A, Midwinter M, Nash GB. Changes in the pattern of plasma extracellular vesicles after severe trauma. PLoS One 2017; 12:e0183640. [PMID: 28837705 PMCID: PMC5570308 DOI: 10.1371/journal.pone.0183640] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 08/08/2017] [Indexed: 11/30/2022] Open
Abstract
Background Extracellular vesicles (EV) released into the circulation after traumatic injury may influence complications. We thus evaluated the numbers of EV in plasma over 28 days after trauma and evaluated their pro-coagulant and inflammatory effects. Methods and findings 37 patients suffering trauma with an injury severity score >15 were studied along with 24 healthy controls. Plasma samples were isolated by double centrifugation (2000g 20min; 13000g 2min) from blood collected from within an hour up to 28 days after injury. Plasma EV were counted and sized using nanoparticle tracking analysis (NTA); counts and cellular origins were also determined by flow cytometry (FC) using cell-specific markers. Functional effects were tested in a procoagulant phospholipid assay and in flow-based, leukocyte adhesion assay after endothelial cells (EC) were treated with EV. We found that EV concentrations measured by NTA were significantly increased in trauma patients compared to healthy controls, and remained elevated over days. In addition, or FC showed that patients with trauma had higher numbers of EV derived from platelets (CD41+), leukocytes (CD45+) and endothelial EC (CD144+). The increases were evident throughout the 28-day follow-up. However, the FC count represented <1% of the count detected by NTA, and only 1–2% of EV identified using NTA had a diameter >400nm. The procoagulant phospholipid activity assay showed that patient plasma accelerated coagulation on day 1 and day 3 after trauma, with coagulation times correlated with EV counts. Furthermore, treatment of EC for 24 hours with plasma containing EV tended to increase the recruitment of peripheral flowing blood mononuclear cells. Conclusions EV counted by FC represent a small sub-population of the total load detected by NTA. Both methods however indicate a significant increase in plasma EV after severe traumatic injury that have pro-coagulant and pro-inflammatory effects that may influence outcomes.
Collapse
Affiliation(s)
- Sahithi J. Kuravi
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- NIHR Surgical Reconstruction and Microbiology Research Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Clara M. Yates
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- NIHR Surgical Reconstruction and Microbiology Research Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Mark Foster
- NIHR Surgical Reconstruction and Microbiology Research Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Paul Harrison
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Jon Hazeldine
- NIHR Surgical Reconstruction and Microbiology Research Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Peter Hampson
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Chris Watson
- Department of Haematology, Queen Elizabeth Hospital, Birmingham, United Kingdom
| | - Antonio Belli
- NIHR Surgical Reconstruction and Microbiology Research Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Mark Midwinter
- NIHR Surgical Reconstruction and Microbiology Research Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Gerard B. Nash
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- NIHR Surgical Reconstruction and Microbiology Research Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
- * E-mail:
| |
Collapse
|
28
|
Herbert JP, Guillotte AR, Hammer RD, Litofsky NS. Coagulopathy in the Setting of Mild Traumatic Brain Injury: Truths and Consequences. Brain Sci 2017; 7:brainsci7070092. [PMID: 28737691 PMCID: PMC5532605 DOI: 10.3390/brainsci7070092] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Revised: 07/09/2017] [Accepted: 07/18/2017] [Indexed: 12/03/2022] Open
Abstract
Mild traumatic brain injury (mTBI) is a common, although poorly-defined clinical entity. Despite its initially mild presentation, patients with mTBI can rapidly deteriorate, often due to significant expansion of intracranial hemorrhage. TBI-associated coagulopathy is the topic of significant clinical and basic science research. Unlike trauma-induced coagulopathy (TIC), TBI-associated coagulopathy does not generally follow widespread injury or global hypoperfusion, suggesting a distinct pathogenesis. Although the fundamental mechanisms of TBI-associated coagulopathy are far from clearly elucidated, several candidate molecules (tissue plasminogen activator (tPA), urokinase plasminogen activator (uPA), tissue factor (TF), and brain-derived microparticles (BDMP)) have been proposed which might explain how even minor brain injury can induce local and systemic coagulopathy. Here, we review the incidence, proposed mechanisms, and common clinical tests relevant to mTBI-associated coagulopathy and briefly summarize our own institutional experience in addition to identifying areas for further research.
Collapse
Affiliation(s)
- Joseph P Herbert
- Division of Neurological Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA.
| | - Andrew R Guillotte
- Division of Neurological Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA.
| | - Richard D Hammer
- Department of Pathology and Anatomical Sciences, University of Missouri School of Medicine, Columbia, MO 65212, USA.
| | - N Scott Litofsky
- Division of Neurological Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA.
| |
Collapse
|
29
|
Zhao Z, Zhou Y, Tian Y, Li M, Dong JF, Zhang J. Cellular microparticles and pathophysiology of traumatic brain injury. Protein Cell 2017; 8:801-810. [PMID: 28466387 PMCID: PMC5676589 DOI: 10.1007/s13238-017-0414-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 04/13/2017] [Indexed: 01/30/2023] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability worldwide. The finding that cellular microparticles (MPs) generated by injured cells profoundly impact on pathological courses of TBI has paved the way for new diagnostic and therapeutic strategies. MPs are subcellular fragments or organelles that serve as carriers of lipids, adhesive receptors, cytokines, nucleic acids, and tissue-degrading enzymes that are unique to the parental cells. Their sub-micron sizes allow MPs to travel to areas that parental cells are unable to reach to exercise diverse biological functions. In this review, we summarize recent developments in identifying a casual role of MPs in the pathologies of TBI and suggest that MPs serve as a new class of therapeutic targets for the prevention and treatment of TBI and associated systemic complications.
Collapse
Affiliation(s)
- Zilong Zhao
- Department of Neurosurgery, Tianjin Institute of Neurology, Tianjin Medical University General Hospital, Tianjin, 300052, China.,BloodWorks Northwest Research Institute, Seattle, WA, 98102, USA
| | - Yuan Zhou
- Department of Neurosurgery, Tianjin Institute of Neurology, Tianjin Medical University General Hospital, Tianjin, 300052, China.,BloodWorks Northwest Research Institute, Seattle, WA, 98102, USA
| | - Ye Tian
- Department of Neurosurgery, Tianjin Institute of Neurology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Min Li
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Jing-Fei Dong
- BloodWorks Northwest Research Institute, Seattle, WA, 98102, USA. .,Division of Hematology, Department of Medicine, School of Medicine, University of Washington, Seattle, WA, 98195, USA.
| | - Jianning Zhang
- Department of Neurosurgery, Tianjin Institute of Neurology, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| |
Collapse
|
30
|
Ochocinska MJ, Zlokovic BV, Searson PC, Crowder AT, Kraig RP, Ljubimova JY, Mainprize TG, Banks WA, Warren RQ, Kindzelski A, Timmer W, Liu CH. NIH workshop report on the trans-agency blood-brain interface workshop 2016: exploring key challenges and opportunities associated with the blood, brain and their interface. Fluids Barriers CNS 2017; 14:12. [PMID: 28457227 PMCID: PMC5410699 DOI: 10.1186/s12987-017-0061-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 04/08/2017] [Indexed: 01/01/2023] Open
Abstract
A trans-agency workshop on the blood–brain interface (BBI), sponsored by the National Heart, Lung and Blood Institute, the National Cancer Institute and the Combat Casualty Care Research Program at the Department of Defense, was conducted in Bethesda MD on June 7–8, 2016. The workshop was structured into four sessions: (1) blood sciences; (2) exosome therapeutics; (3) next generation in vitro blood–brain barrier (BBB) models; and (4) BBB delivery and targeting. The first day of the workshop focused on the physiology of the blood and neuro-vascular unit, blood or biofluid-based molecular markers, extracellular vesicles associated with brain injury, and how these entities can be employed to better evaluate injury states and/or deliver therapeutics. The second day of the workshop focused on technical advances in in vitro models, BBB manipulations and nanoparticle-based drug carrier designs, with the goal of improving drug delivery to the central nervous system. The presentations and discussions underscored the role of the BBI in brain injury, as well as the role of the BBB as both a limiting factor and a potential conduit for drug delivery to the brain. At the conclusion of the meeting, the participants discussed challenges and opportunities confronting BBI translational researchers. In particular, the participants recommended using BBI translational research to stimulate advances in diagnostics, as well as targeted delivery approaches for detection and therapy of both brain injury and disease.
Collapse
Affiliation(s)
- Margaret J Ochocinska
- National Heart, Lung, and Blood Institute, National Institutes of Health, 6701 Rockledge Dr., Room 9149, Bethesda, MD, 20892-7950, USA.
| | | | | | | | | | | | | | | | - Ronald Q Warren
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Andrei Kindzelski
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - William Timmer
- National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Christina H Liu
- National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
31
|
Kumar A, Stoica BA, Loane DJ, Yang M, Abulwerdi G, Khan N, Kumar A, Thom SR, Faden AI. Microglial-derived microparticles mediate neuroinflammation after traumatic brain injury. J Neuroinflammation 2017; 14:47. [PMID: 28292310 PMCID: PMC5351060 DOI: 10.1186/s12974-017-0819-4] [Citation(s) in RCA: 239] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 02/18/2017] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Local and systemic inflammatory responses are initiated early after traumatic brain injury (TBI), and may play a key role in the secondary injury processes resulting in neuronal loss and neurological deficits. However, the mechanisms responsible for the rapid expansion of neuroinflammation and its long-term progression have yet to be elucidated. Here, we investigate the role of microparticles (MP), a member of the extracellular vesicle family, in the exchange of pro-inflammatory molecules between brain immune cells, as well as their transfer to the systemic circulation, as key pathways of inflammation propagation following brain trauma. METHODS Adult male C57BL/6 mice were subjected to controlled cortical impact TBI for 24 h, and enriched MP were isolated in the blood, while neuroinflammation was assessed in the TBI cortex. MP were characterized by flow cytometry, and MP content was assayed using gene and protein markers for pro-inflammatory mediators. Enriched MP co-cultured with BV2 or primary microglial cells were used for immune propagation assays. Enriched MP from BV2 microglia or CD11b-positive microglia from the TBI brain were stereotactically injected into the cortex of uninjured mice to evaluate MP-related seeding of neuroinflammation in vivo. RESULTS As the neuroinflammatory response is developing in the brain after TBI, microglial-derived MP are released into the circulation. Circulating enriched MP from the TBI animals can activate microglia in vitro. Lipopolysaccharide stimulation increases MP release from microglia in vitro and enhances their content of pro-inflammatory mediators, interleukin-1β and microRNA-155. Enriched MP from activated microglia in vitro or CD11b-isolated microglia/macrophage from the TBI brain ex vivo are sufficient to initiate neuroinflammation following their injection into the cortex of naïve (uninjured) animals. CONCLUSIONS These data provide further insights into the mechanisms underlying the development and dissemination of neuroinflammation after TBI. MP loaded with pro-inflammatory molecules initially released by microglia following trauma can activate additional microglia that may contribute to progressive neuroinflammatory response in the injured brain, as well as stimulate systemic immune responses. Due to their ability to independently initiate inflammatory responses, MP derived from activated microglia may provide a potential therapeutic target for other neurological disorders in which neuroinflammation may be a contributing factor.
Collapse
Affiliation(s)
- Alok Kumar
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD, USA.,Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Health Sciences Facility II (HSFII), #S247 20 Penn Street, Baltimore, MD, 21201, USA
| | - Bogdan A Stoica
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD, USA.,Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Health Sciences Facility II (HSFII), #S247 20 Penn Street, Baltimore, MD, 21201, USA
| | - David J Loane
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD, USA.,Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Health Sciences Facility II (HSFII), #S247 20 Penn Street, Baltimore, MD, 21201, USA
| | - Ming Yang
- Department of Emergency Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Gelareh Abulwerdi
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD, USA.,Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Health Sciences Facility II (HSFII), #S247 20 Penn Street, Baltimore, MD, 21201, USA
| | - Niaz Khan
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD, USA.,Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Health Sciences Facility II (HSFII), #S247 20 Penn Street, Baltimore, MD, 21201, USA
| | - Asit Kumar
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD, USA.,Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Health Sciences Facility II (HSFII), #S247 20 Penn Street, Baltimore, MD, 21201, USA
| | - Stephen R Thom
- Department of Emergency Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Alan I Faden
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD, USA. .,Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Health Sciences Facility II (HSFII), #S247 20 Penn Street, Baltimore, MD, 21201, USA.
| |
Collapse
|
32
|
Vanderwerf JD, Kumar MA. Management of neurologic complications of coagulopathies. HANDBOOK OF CLINICAL NEUROLOGY 2017; 141:743-764. [PMID: 28190445 DOI: 10.1016/b978-0-444-63599-0.00040-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Coagulopathy is common in intensive care units (ICUs). Many physiologic derangements lead to dysfunctional hemostasis; these may be either congenital or acquired. The most devastating outcome of coagulopathy in the critically ill is major bleeding, defined by transfusion requirement, hemodynamic instability, or intracranial hemorrhage. ICU coagulopathy often poses complex management dilemmas, as bleeding risk must be tempered with thrombotic potential. Coagulopathy associated with intracranial hemorrhage bears directly on prognosis and outcome. There is a paucity of high-quality evidence for the management of coagulopathies in neurocritical care; however, data derived from studies of patients with intraparenchymal hemorrhage may inform treatment decisions. Coagulopathy is often broadly defined as any derangement of hemostasis resulting in either excessive bleeding or clotting, although most typically it is defined as impaired clot formation. Abnormalities in coagulation testing without overt clinical bleeding may also be considered evidence of coagulopathy. This chapter will focus on acquired conditions, such as organ failure, pharmacologic therapies, and platelet dysfunction that are associated with defective clot formation and result in, or exacerbate, intracranial hemorrhage, specifically spontaneous intraparenchymal hemorrhage and traumatic brain injury.
Collapse
Affiliation(s)
- J D Vanderwerf
- Department of Neurology, Perelman School of Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - M A Kumar
- Departments of Neurology, Neurosurgery, Anesthesiology and Critical Care, Perelman School of Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
33
|
O’Dea KP, Porter JR, Tirlapur N, Katbeh U, Singh S, Handy JM, Takata M. Circulating Microvesicles Are Elevated Acutely following Major Burns Injury and Associated with Clinical Severity. PLoS One 2016; 11:e0167801. [PMID: 27936199 PMCID: PMC5148002 DOI: 10.1371/journal.pone.0167801] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 11/21/2016] [Indexed: 01/26/2023] Open
Abstract
Microvesicles are cell-derived signaling particles emerging as important mediators and biomarkers of systemic inflammation, but their production in severe burn injury patients has not been described. In this pilot investigation, we measured circulating microvesicle levels following severe burns, with severe sepsis patients as a comparator group. We hypothesized that levels of circulating vascular cell-derived microvesicles are elevated acutely following burns injury, mirroring clinical severity due to the early onset and prevalence of systemic inflammatory response syndrome (SIRS) in these patients. Blood samples were obtained from patients with moderate to severe thermal injury burns, with severe sepsis, and from healthy volunteers. Circulating microvesicles derived from total leukocytes, granulocytes, monocytes, and endothelial cells were quantified in plasma by flow cytometry. All circulating microvesicle subpopulations were elevated in burns patients on day of admission (day 0) compared to healthy volunteers (leukocyte-microvesicles: 3.5-fold, p = 0.005; granulocyte-microvesicles: 12.8-fold, p<0.0001; monocyte-microvesicles: 20.4-fold, p<0.0001; endothelial- microvesicles: 9.6-fold, p = 0.01), but decreased significantly by day 2. Microvesicle levels were increased with severe sepsis, but less consistently between patients. Leukocyte- and granulocyte-derived microvesicles on day 0 correlated with clinical assessment scores and were higher in burns ICU non-survivors compared to survivors (leukocyte MVs 4.6 fold, p = 0.002; granulocyte MVs 4.8 fold, p = 0.003). Mortality prediction analysis of area under receiver operating characteristic curve was 0.92 (p = 0.01) for total leukocyte microvesicles and 0.85 (p = 0.04) for granulocyte microvesicles. These findings demonstrate, for the first time, acute increases in circulating microvesicles following burns injury in patients and point to their potential role in propagation of sterile SIRS-related pathophysiology.
Collapse
Affiliation(s)
- Kieran P. O’Dea
- Section of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery & Cancer, Imperial College London, London, United Kingdom
| | - John R. Porter
- Section of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery & Cancer, Imperial College London, London, United Kingdom
- Intensive Care Unit, Chelsea and Westminster Hospital, London, United Kingdom
| | - Nikhil Tirlapur
- Section of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery & Cancer, Imperial College London, London, United Kingdom
| | - Umar Katbeh
- Section of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery & Cancer, Imperial College London, London, United Kingdom
| | - Suveer Singh
- Section of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery & Cancer, Imperial College London, London, United Kingdom
- Intensive Care Unit, Chelsea and Westminster Hospital, London, United Kingdom
| | - Jonathan M. Handy
- Section of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery & Cancer, Imperial College London, London, United Kingdom
- Intensive Care Unit, Chelsea and Westminster Hospital, London, United Kingdom
| | - Masao Takata
- Section of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery & Cancer, Imperial College London, London, United Kingdom
| |
Collapse
|
34
|
Yun JW, Xiao A, Tsunoda I, Minagar A, Alexander JS. From trash to treasure: The untapped potential of endothelial microparticles in neurovascular diseases. PATHOPHYSIOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY FOR PATHOPHYSIOLOGY 2016; 23:265-274. [PMID: 27531185 DOI: 10.1016/j.pathophys.2016.08.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 08/04/2016] [Accepted: 08/12/2016] [Indexed: 02/08/2023]
Abstract
Discovered in 1947, microparticles (MP) represent a group of sub-micron cell-derived particles isolated by high speed centrifugation. Once regarded as cellular 'trash', in the past decade MP have gained tremendous attention in both basic sciences and medical research both as biomarkers and mediators of infection, injury and response to therapy. Because MP bear cell surface markers derived from parent cells, accumulate in extracellular fluids (plasma, serum, milk, urine, cerebrospinal fluid) MP based tests are being developed commercially as important components in 'liquid biopsy' approaches, providing valuable readouts in cardiovascular disease and cancer, as well as stroke, Alzheimer's disease and Multiple Sclerosis. Importantly, MP have been reported as mobile transport vectors in the intercellular transfer of mRNAs, microRNAs, lipids and proteins. Here we discuss MP structure, properties and functions with particular relevance to neurological and neurovascular diseases.
Collapse
Affiliation(s)
- J Winny Yun
- Departments of Molecular & Cellular Physiology, LSU Health Sciences Center, Shreveport, LA, United States
| | - Adam Xiao
- Departments of Molecular & Cellular Physiology, LSU Health Sciences Center, Shreveport, LA, United States
| | - Ikuo Tsunoda
- Departments of Neurology, LSU Health Sciences Center, Shreveport, LA, United States; Department of Microbiology, Kindai University, Faculty of Medicine, 377-2 Ohnohigashi, Osakasayama, Osaka, 589-8511, Japan
| | - Alireza Minagar
- Departments of Neurology, LSU Health Sciences Center, Shreveport, LA, United States
| | - J Steven Alexander
- Departments of Molecular & Cellular Physiology, LSU Health Sciences Center, Shreveport, LA, United States; Departments of Neurology, LSU Health Sciences Center, Shreveport, LA, United States.
| |
Collapse
|
35
|
Advances in the understanding of trauma-induced coagulopathy. Blood 2016; 128:1043-9. [PMID: 27381903 DOI: 10.1182/blood-2016-01-636423] [Citation(s) in RCA: 191] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 06/27/2016] [Indexed: 01/10/2023] Open
Abstract
Ten percent of deaths worldwide are due to trauma, and it is the third most common cause of death in the United States. Despite a profound upregulation in procoagulant mechanisms, one-quarter of trauma patients present with laboratory-based evidence of trauma-induced coagulopathy (TIC), which is associated with poorer outcomes including increased mortality. The most common causes of death after trauma are hemorrhage and traumatic brain injury (TBI). The management of TIC has significant implications in both because many hemorrhagic deaths could be preventable, and TIC is associated with progression of intracranial injury after TBI. This review covers the most recent evidence and advances in our understanding of TIC, including the role of platelet dysfunction, endothelial activation, and fibrinolysis. Trauma induces a plethora of biochemical and physiologic changes, and despite numerous studies reporting differences in coagulation parameters between trauma patients and uninjured controls, it is unclear whether some of these differences may be "normal" after trauma. Comparisons between trauma patients with differing outcomes and use of animal studies have shed some light on this issue, but much of the data continue to be correlative with causative links lacking. In particular, there are little data linking the laboratory-based abnormalities with true clinically evident coagulopathic bleeding. For these reasons, TIC continues to be a significant diagnostic and therapeutic challenge.
Collapse
|
36
|
Abstract
All known cells continuously release nanoscale lipid membrane-enclosed packets. These packets, termed extracellular vesicles (EVs), bear the signature of their cells of origin. These vesicles can be detected in just about every type of biofluid tested, including blood, urine, and cerebrospinal fluid. The majority comes from normal cells, but disease cells also release them. There is a great interest in collecting and analyzing EVs in biofluids as diagnostics for a wide spectrum of central nervous system diseases. Here, we will review the state of central nervous system EV research in terms of molecular diagnostics and biomarkers.
Collapse
|
37
|
Yasui H, Donahue DL, Walsh M, Castellino FJ, Ploplis VA. Early coagulation events induce acute lung injury in a rat model of blunt traumatic brain injury. Am J Physiol Lung Cell Mol Physiol 2016; 311:L74-86. [PMID: 27190065 PMCID: PMC4967191 DOI: 10.1152/ajplung.00429.2015] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 05/17/2016] [Indexed: 12/20/2022] Open
Abstract
Acute lung injury (ALI) and systemic coagulopathy are serious complications of traumatic brain injury (TBI) that frequently lead to poor clinical outcomes. Although the release of tissue factor (TF), a potent initiator of the extrinsic pathway of coagulation, from the injured brain is thought to play a key role in coagulopathy after TBI, its function in ALI following TBI remains unclear. In this study, we investigated whether the systemic appearance of TF correlated with the ensuing coagulopathy that follows TBI in ALI using an anesthetized rat blunt trauma TBI model. Blood and lung samples were obtained after TBI. Compared with controls, pulmonary edema and increased pulmonary permeability were observed as early as 5 min after TBI without evidence of norepinephrine involvement. Systemic TF increased at 5 min and then diminished 60 min after TBI. Lung injury and alveolar hemorrhaging were also observed as early as 5 min after TBI. A biphasic elevation of TF was observed in the lungs after TBI, and TF-positive microparticles (MPs) were detected in the alveolar spaces. Fibrin(ogen) deposition was also observed in the lungs within 60 min after TBI. Additionally, preadministration of a direct thrombin inhibitor, Refludan, attenuated lung injuries, thus implicating thrombin as a direct participant in ALI after TBI. The results from this study demonstrated that enhanced systemic TF may be an initiator of coagulation activation that contributes to ALI after TBI.
Collapse
Affiliation(s)
- Hideki Yasui
- W. M. Keck Center for Transgene Research, University of Notre Dame, Notre Dame, Indiana
| | - Deborah L Donahue
- W. M. Keck Center for Transgene Research, University of Notre Dame, Notre Dame, Indiana
| | - Mark Walsh
- W. M. Keck Center for Transgene Research, University of Notre Dame, Notre Dame, Indiana; Memorial Hospital of South Bend, South Bend, Indiana
| | - Francis J Castellino
- W. M. Keck Center for Transgene Research, University of Notre Dame, Notre Dame, Indiana; Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana; and
| | - Victoria A Ploplis
- W. M. Keck Center for Transgene Research, University of Notre Dame, Notre Dame, Indiana; Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana; and
| |
Collapse
|
38
|
Lacedonia D, Carpagnano GE, Trotta T, Palladino GP, Panaro MA, Zoppo LD, Foschino Barbaro MP, Porro C. Microparticles in sputum of COPD patients: a potential biomarker of the disease? Int J Chron Obstruct Pulmon Dis 2016; 11:527-33. [PMID: 27042041 PMCID: PMC4798204 DOI: 10.2147/copd.s99547] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Background Microparticles (MPs) are small membrane vesicles of 0.1–1 µm which are released by cells following chemical, physical, and apoptotic stimuli. MPs represent more than a miniature version of the cell. Their composition and function depend not only on cellular origin, but also on stimuli. Chronic obstructive pulmonary disease (COPD) is a lung disease characterized by nearly irreversible lung destruction which results in airway limitation. Purpose We investigated the presence and source of MPs in sputum of COPD patients to evaluate if changes in MP number and origin may reflect the pathophysiological conditions of disease and may serve as potential biomarkers for diagnostic and prognostic use. Methods Induced sputum samples were collected from 18 male subjects and liquefied with Sputasol. MPs obtained were immunolabeled for leukocyte (CD11a), granulocyte (CD66b), monocyte-macrophage (CD11b), platelets and megakaryocytic cells (CD41), endothelial cells (CD31), and red blood cells (CD235ab) and analyzed by cytofluorimetry. Results There was a negative correlation between CD31-MPs and forced expiratory volume in 1 second (R=−53, P<0.05) and CD66b-MP level was correlated with worse performance index of COPD such as the Body mass index airflow Obstruction, Dyspnea, and Exercise capacity (BODE); they were negatively correlated with 6-minute walking test: 0.65 and −0.64, respectively (P<0.05). CD235ab-MPs showed a negative correlation with body mass index (R=−0.86, P<0.05), while there was a positive correlation with dyspnea index (R=0.91, P<0.05). Conclusion The main finding of this study was that MPs were detected in the sputum of patients affected by COPD. The phenotype of some of them was related to the main COPD parameters. These results suggest that MPs could be implicated in the pathogenesis of COPD.
Collapse
Affiliation(s)
- Donato Lacedonia
- Department of Medical and Surgical Sciences, Institute of Respiratory Diseases, University of Foggia, Foggia, Italy
| | - Giovanna Elisiana Carpagnano
- Department of Medical and Surgical Sciences, Institute of Respiratory Diseases, University of Foggia, Foggia, Italy
| | - Teresa Trotta
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Grazia Pia Palladino
- Department of Medical and Surgical Sciences, Institute of Respiratory Diseases, University of Foggia, Foggia, Italy
| | - Maria Antonietta Panaro
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Liugi Davide Zoppo
- Department of Medical and Surgical Sciences, Institute of Respiratory Diseases, University of Foggia, Foggia, Italy
| | - Maria Pia Foschino Barbaro
- Department of Medical and Surgical Sciences, Institute of Respiratory Diseases, University of Foggia, Foggia, Italy
| | - Chiara Porro
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| |
Collapse
|
39
|
Matijevic N, Wang YWW, Holcomb JB, Kozar R, Cardenas JC, Wade CE. Microvesicle phenotypes are associated with transfusion requirements and mortality in subjects with severe injuries. J Extracell Vesicles 2015; 4:29338. [PMID: 26689982 PMCID: PMC4685295 DOI: 10.3402/jev.v4.29338] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 11/23/2015] [Accepted: 11/26/2015] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Severe injury often results in substantial bleeding and mortality. Injury provokes cellular activation and release of extracellular vesicles. Circulating microvesicles (MVs) are predominantly platelet-derived and highly procoagulant. They support hemostasis and vascular function. The roles of MVs in survival after severe injury are largely unknown. We hypothesized that altered MV phenotypes would be associated with transfusion requirements and poor outcomes. METHODS This single-centre study was approved by the Institutional Review Board. The study cohort consisted of patients with major trauma requiring blood product transfusion and 26 healthy controls. Plasma samples for MVs were collected upon admission to the emergency department (n=169) and post-resuscitation (n=42), and analysed by flow cytometry for MV counts and cellular origin: platelet (PMV), erythrocyte (RMV), leukocyte (LMV), endothelial (EMV), tissue factor (TFMV), and annexin V (AVMV). Twenty-four hour mortality is the outcome measurement used to classify survivors versus non-survivors. Data were compared over time and analysed with demographic and clinical data. RESULTS The median age was 34 (IQR 23, 51), 72% were male, Injury Severity Score was 29 (IQR 19, 36), and 24 h mortality was 13%. MV levels and phenotypes differed between patients and controls. Elevated admission EMVs were found both in survivors (409/µL) and non-survivors (393/µL) compared to controls (23/µL, p<0.001) and persisted over time. Admission levels of PMV, AVMV, RMV, and TFMV were significantly lower in patients who died compared to survivors, but were not independently associated with the 24 h mortality rate. Patients with low MV levels at admission received the most blood products within the first 24 h. AVMV and PMV levels either increased over time or stabilized in survivors but decreased in non-survivors, resulting in significantly lower levels at intensive care unit admission in non-survivors (1,048 vs. 1,880 AVMV/µL, p<0.00004 and 1,245 PMP/µL vs. 1,866 PMP/µL, p=0.003). CONCLUSION Severe injury results in endothelial activation and altered MV phenotypes. Significant differences in specific MV phenotypes or changes over time were associated with blood product requirements and the 24 h mortality rate.
Collapse
Affiliation(s)
- Nena Matijevic
- Department of Surgery, The University of Texas Health Science Center at Houston, Houston, TX, USA.,The Center for Translational Injury Research, The University of Texas Health Science Center at Houston, Houston, TX, USA;
| | - Yao-Wei W Wang
- The Center for Translational Injury Research, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - John B Holcomb
- Department of Surgery, The University of Texas Health Science Center at Houston, Houston, TX, USA.,The Center for Translational Injury Research, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Rosemary Kozar
- Department of Surgery, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jessica C Cardenas
- Department of Surgery, The University of Texas Health Science Center at Houston, Houston, TX, USA.,The Center for Translational Injury Research, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Charles E Wade
- Department of Surgery, The University of Texas Health Science Center at Houston, Houston, TX, USA.,The Center for Translational Injury Research, The University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
40
|
Bohman LE, Riley J, Milovanova TN, Sanborn MR, Thom SR, Armstead WM. Microparticles Impair Hypotensive Cerebrovasodilation and Cause Hippocampal Neuronal Cell Injury after Traumatic Brain Injury. J Neurotrauma 2015; 33:168-74. [PMID: 26230045 DOI: 10.1089/neu.2015.3885] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Endothelin-1 (ET-1), tissue plasminogen activator (tPA), and extracellular signal-regulated kinases-mitogen activated protein kinase (ERK-MAPK) are mediators of impaired cerebral hemodynamics after fluid percussion brain injury (FPI) in piglets. Microparticles (MPs) are released into the circulation from a variety of cells during stress, are pro-thrombotic and pro-inflammatory, and may be lysed with polyethylene glycol telomere B (PEG-TB). We hypothesized that MPs released after traumatic brain injury impair hypotensive cerebrovasodilation and that PEG-TB protects the vascular response via MP lysis, and we investigated the relationship between MPs, tPA, ET-1, and ERK-MAPK in that process. FPI was induced in piglets equipped with a closed cranial window. Animals received PEG-TB or saline (vehicle) 30-minutes post-injury. Serum and cerebrospinal fluid (CSF) were sampled and pial arteries were measured pre- and post-injury. MPs were quantified by flow cytometry. CSF samples were analyzed with enzyme-linked immunosorbent assay. MP levels, vasodilatory responses, and CSF signaling assays were similar in all animals prior to injury and treatment. After injury, MP levels were elevated in the serum of vehicle but not in PEG-TB-treated animals. Pial artery dilation in response to hypotension was impaired after injury but protected in PEG-TB-treated animals. After injury, CSF levels of tPA, ET-1, and ERK-MAPK were all elevated, but not in PEG-TB-treated animals. PEG-TB-treated animals also showed reduction in neuronal injury in CA1 and CA3 hippocampus, compared with control animals. These results show that serum MP levels are elevated after FPI and lead to impaired hypotensive cerebrovasodilation via over-expression of tPA, ET-1, and ERK-MAPK. Treatment with PEG-TB after injury reduces MP levels and protects hypotensive cerebrovasodilation and limits hippocampal neuronal cell injury.
Collapse
Affiliation(s)
- Leif-Erik Bohman
- 1 Department of Neurosurgery, University of Pennsylvania , Philadelphia, Pennsylvania
| | - John Riley
- 2 Department of Anesthesiology and Critical Care, University of Pennsylvania , Philadelphia, Pennsylvania
| | - Tatyana N Milovanova
- 3 Department of Emergency Medicine, University of Pennsylvania , Philadelphia, Pennsylvania.,5 Institute for Environmental Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
| | - Matthew R Sanborn
- 1 Department of Neurosurgery, University of Pennsylvania , Philadelphia, Pennsylvania
| | - Stephen R Thom
- 3 Department of Emergency Medicine, University of Pennsylvania , Philadelphia, Pennsylvania.,5 Institute for Environmental Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
| | - William M Armstead
- 2 Department of Anesthesiology and Critical Care, University of Pennsylvania , Philadelphia, Pennsylvania.,4 Department of Pharmacology, University of Pennsylvania , Philadelphia, Pennsylvania
| |
Collapse
|
41
|
Evolving beyond the vicious triad: Differential mediation of traumatic coagulopathy by injury, shock, and resuscitation. J Trauma Acute Care Surg 2015; 78:516-23. [PMID: 25710421 DOI: 10.1097/ta.0000000000000545] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND A subset of trauma patients with critical injury present with coagulopathy, portending markedly worse outcomes. Clinical practice is evolving to treat the classical risk factors of hypothermia, hemodilution, and acidosis; however, coagulopathy persists even in the absence of these factors. We sought to determine the relative importance of injury- and shock-specific factors compared with resuscitation-associated factors in coagulopathy after trauma. METHODS Comprehensive demographic data, laboratory data, and outcomes data were prospectively collected from seven trauma centers over 8 years (November 2003 to August 2011) as part of the Inflammation and the Host Response to Injury Large-Scale Collaborative Program. A total of 1,537 critically injured patients with blunt trauma and hemorrhagic shock were analyzed to evaluate predictors of admission coagulopathy (international normalized ratio [INR] ≥ 1.3), multiorgan failure, and mortality. RESULTS Of 1,537 patients, 578 (37.6%) had admission INR of 1.3 or greater. Coagulopathic patients had more severe injury, more severe base deficit and lactate levels, as well as lower admission temperature, lower pH, and higher prehospital crystalloid volume (all p < 0.001). Coagulopathic patients required more blood products and mechanical ventilation and had higher rates of nosocomial infection, multiorgan failure, and mortality (all p < 0.02). Injury severity, temperature, and acidosis (all p < 0.02) independently predicted coagulopathy in multivariate analysis, with a significant interaction between lactate and prehospital crystalloid. In Cox regression models, however, coagulopathy itself remained an independent predictor of both multiorgan failure and mortality (p < 0.02) even when adjusted for injury severity, shock, and elements of the vicious triad. CONCLUSION Most patients with coagulopathy after trauma have mixed risk factors; however, coagulopathy has deleterious effects independent of injury severity, shock, and the vicious triad. Better understanding of the biochemical mechanisms of acute traumatic coagulopathy may facilitate biochemically targeted resuscitation strategies and improve outcomes. LEVEL OF EVIDENCE Prognostic and epidemiologic study, level II.
Collapse
|
42
|
Parida BK, Garrastazu H, Aden JK, Cap AP, McFaul SJ. Silica microspheres are superior to polystyrene for microvesicle analysis by flow cytometry. Thromb Res 2015; 135:1000-6. [DOI: 10.1016/j.thromres.2015.02.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 01/21/2015] [Accepted: 02/10/2015] [Indexed: 01/06/2023]
|
43
|
Tian Y, Salsbery B, Wang M, Yuan H, Yang J, Zhao Z, Wu X, Zhang Y, Konkle BA, Thiagarajan P, Li M, Zhang J, Dong JF. Brain-derived microparticles induce systemic coagulation in a murine model of traumatic brain injury. Blood 2015; 125:2151-9. [PMID: 25628471 PMCID: PMC4375111 DOI: 10.1182/blood-2014-09-598805] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 01/12/2015] [Indexed: 12/14/2022] Open
Abstract
Traumatic brain injury (TBI) is associated with coagulopathy, although it often lacks 2 key risk factors: severe bleeding and significant fluid resuscitation associated with hemorrhagic shock. The pathogenesis of TBI-associated coagulopathy remains poorly understood. We tested the hypothesis that brain-derived microparticles (BDMPs) released from an injured brain induce a hypercoagulable state that rapidly turns into consumptive coagulopathy. Here, we report that mice subjected to fluid percussion injury (1.9 ± 0.1 atm) developed a BDMP-dependent hypercoagulable state, with peak levels of plasma glial cell and neuronal BDMPs reaching 17 496 ± 4833/μL and 18 388 ± 3657/μL 3 hours after TBI, respectively. Uninjured mice injected with BDMPs developed a dose-dependent hyper-turned hypocoagulable state measured by a progressively prolonged clotting time, fibrinogen depletion, and microvascular fibrin deposition in multiple organs. The BDMPs were 50 to 300 nm with intact membranes, expressing neuronal or glial cell markers and procoagulant phosphatidylserine and tissue factor. Their procoagulant activity was greater than platelet microparticles and was dose-dependently blocked by lactadherin. Microparticles were produced from injured hippocampal cells, transmigrated through the disrupted endothelial barrier in a platelet-dependent manner, and activated platelets. These data define a novel mechanism of TBI-associated coagulopathy in mice, identify early predictive markers, and provide alternative therapeutic targets.
Collapse
Affiliation(s)
- Ye Tian
- Tianjin Neurological Institute, Department of Neurosurgery, General Hospital, Tianjin Medical University, Tianjin, China; Puget Sound Blood Research Institute, Seattle, WA
| | | | - Min Wang
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Hengjie Yuan
- Tianjin Neurological Institute, Department of Neurosurgery, General Hospital, Tianjin Medical University, Tianjin, China; Puget Sound Blood Research Institute, Seattle, WA
| | - Jing Yang
- Puget Sound Blood Research Institute, Seattle, WA
| | - Zilong Zhao
- Tianjin Neurological Institute, Department of Neurosurgery, General Hospital, Tianjin Medical University, Tianjin, China
| | - Xiaoping Wu
- Puget Sound Blood Research Institute, Seattle, WA
| | - Yanjun Zhang
- Tianjin Neurological Institute, Department of Neurosurgery, General Hospital, Tianjin Medical University, Tianjin, China
| | - Barbara A Konkle
- Puget Sound Blood Research Institute, Seattle, WA; Division of Hematology, Department of Medicine, University of Washington School of Medicine, Seattle, WA
| | - Perumal Thiagarajan
- Departments of Pathology and Medicine, Baylor College of Medicine, Houston, TX; and Center for Translational Research on Inflammatory Diseases, Michael E. DeBakey VA Medical Center, Houston, TX
| | - Min Li
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Jianning Zhang
- Tianjin Neurological Institute, Department of Neurosurgery, General Hospital, Tianjin Medical University, Tianjin, China
| | - Jing-Fei Dong
- Puget Sound Blood Research Institute, Seattle, WA; Division of Hematology, Department of Medicine, University of Washington School of Medicine, Seattle, WA
| |
Collapse
|
44
|
Midura EF, Jernigan PL, Kuethe JW, Friend LA, Veile R, Makley AT, Caldwell CC, Goodman MD. Microparticles impact coagulation after traumatic brain injury. J Surg Res 2015; 197:25-31. [PMID: 25846728 DOI: 10.1016/j.jss.2015.02.064] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 02/18/2015] [Accepted: 02/26/2015] [Indexed: 02/06/2023]
Abstract
BACKGROUND The pathophysiology that drives the subacute hypercoagulable state commonly seen after traumatic brain injury (TBI) is not well understood. Alterations caused by TBI in platelet and microparticle (MP) numbers and function have been suggested as possible causes; however, the contributions of platelets and MPs are currently unknown. MATERIALS AND METHODS A weight-drop technique of TBI using a murine model of moderate head injury was used. Blood was collected at intervals after injury. MP enumeration and characterization were performed using Nanoparticle Tracking Analysis, and platelet counts and coagulation parameters were determined using thromboelastometry. A MP procoagulant assay was used to compare activity between injured and sham mice. RESULTS At 24 h after injury, there were no changes in circulating platelet numbers. However, there was a decrease in platelet contribution to clot formation. In contrast, there was a decline in circulating total MP numbers. When MPs from sham mice were added to the blood from head-injured animals, there was a normalization of platelet contribution to clot formation. Conversely, when MPs from TBI mice were added to sham blood, there was a significant decrease in platelet contribution to clot formation. Notably, there was an increase in MP procoagulant activity in head-injured mice. CONCLUSIONS MPs generated after TBI likely contribute to altered coagulation after head injury and may play a key role in the development of a posttraumatic hypercoagulable state in TBI patients.
Collapse
Affiliation(s)
- Emily F Midura
- Division of Research, Department of Surgery and Institute for Military Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Peter L Jernigan
- Division of Research, Department of Surgery and Institute for Military Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Joshua W Kuethe
- Division of Research, Department of Surgery and Institute for Military Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Lou Ann Friend
- Division of Research, Department of Surgery and Institute for Military Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Rosalie Veile
- Division of Research, Department of Surgery and Institute for Military Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Amy T Makley
- Division of Research, Department of Surgery and Institute for Military Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Charles C Caldwell
- Division of Research, Department of Surgery and Institute for Military Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Michael D Goodman
- Division of Research, Department of Surgery and Institute for Military Medicine, University of Cincinnati, Cincinnati, Ohio.
| |
Collapse
|
45
|
Mooberry MJ, Key NS. Microparticle analysis in disorders of hemostasis and thrombosis. Cytometry A 2015; 89:111-22. [PMID: 25704723 DOI: 10.1002/cyto.a.22647] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Revised: 01/29/2015] [Accepted: 02/03/2015] [Indexed: 01/18/2023]
Abstract
Microparticles (MPs) are submicron vesicles released from the plasma membrane of eukaryotic cells in response to activation or apoptosis. MPs are known to be involved in numerous biologic processes, including inflammation, the immune response, cancer metastasis, and angiogenesis. Their earliest recognized and most widely accepted role, however, is the ability to promote and support the process of blood coagulation. Consequently, there is ongoing interest in studying MPs in disorders of hemostasis and thrombosis. Both phosphatidylserine (PS) exposure and the presence of tissue factor (TF) in the MP membrane may account for their procoagulant properties, and elevated numbers of MPs in plasma have been reported in numerous prothrombotic conditions. To date, however, there are few data on true causality linking MPs to the genesis of thrombosis. A variety of methodologies have been employed to characterize and quantify MPs, although detection is challenging due to their submicron size. Flow cytometry (FCM) remains the most frequently utilized strategy for MP detection; however, it is associated with significant technological limitations. Additionally, preanalytical and analytical variables can influence the detection of MPs by FCM, rendering data interpretation difficult. Lack of methodologic standardization in MP analysis by FCM confounds the issue further, although efforts are currently underway to address this limitation. Moving forward, it will be important to address these technical challenges as a scientific community if we are to better understand the role that MPs play in disorders of hemostasis and thrombosis.
Collapse
Affiliation(s)
- Micah J Mooberry
- Department of Medicine, Division of Hematology/Oncology, University of North Carolina at Chapel Hill, North Carolina
| | - Nigel S Key
- Department of Medicine, Division of Hematology/Oncology, University of North Carolina at Chapel Hill, North Carolina
| |
Collapse
|
46
|
van Ierssel SH, Conraads VM, Van Craenenbroeck EM, Liu Y, Maas AIR, Parizel PM, Hoymans VY, Vrints CJ, Jorens PG. Endothelial dysfunction in acute brain injury and the development of cerebral ischemia. J Neurosci Res 2015; 93:866-72. [PMID: 25677574 DOI: 10.1002/jnr.23566] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2014] [Revised: 11/22/2014] [Accepted: 01/05/2015] [Indexed: 01/13/2023]
Abstract
Cerebral ischemia (CeI) is a major complicating event after acute brain injury (ABI) in which endothelial dysfunction is a key player. This study evaluates cellular markers of endothelial function and in vivo reactive hyperemia in patients with ABI and their relationship to the development of cerebral ischemia. We studied cellular markers of endothelial dysfunction and the peripheral reactive hyperemia index (RHI) in 26 patients with ABI at admission and after 6 and 12 days, and compared these with those of healthy volunteers (n = 15). CeI was determined clinically or by computer tomography. In patients with ABI, RHI at admission was significantly reduced compared with healthy subjects (P = 0.003), coinciding with a decrease in circulating endothelial progenitor cells (EPC; P = 0.002). The RHI recovered in eight patients without development of CeI, but failed to fully recover by day 12 in three of four patients who developed CeI. Despite recovery of the RHI within 12 days in these patients (P = 0.003), EPC count remained significantly lower after 12 days in patients with ABI (P = 0.022). CD31(+) T cells and endothelial microparticles were not different between controls and patients. No differences were noted in cellular markers of endothelial dysfunction in patients developing CeI and those not. In conclusion, patients with ABI exhibit impaired microvascular endothelial function measured as RHI and a decreased circulating level of EPC.
Collapse
Affiliation(s)
- Sabrina H van Ierssel
- Department of Critical Care Medicine, Antwerp University Hospital, University of Antwerp, Edegem, Belgium; Department of Internal Medicine, Antwerp University Hospital, University of Antwerp, Edegem, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Cap A, Hunt BJ. The pathogenesis of traumatic coagulopathy. Anaesthesia 2015; 70 Suppl 1:96-101, e32-4. [PMID: 25440402 DOI: 10.1111/anae.12914] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/19/2014] [Indexed: 12/14/2022]
Abstract
Over the last 10 years, the management of major haemorrhage in trauma patients has changed radically. This is mainly due to the recognition that many patients who are bleeding when they come in to the emergency department have an established coagulopathy before the haemodilution effects of fluid resuscitation. This has led to the use of new terminology: acute traumatic coagulopathy, acute coagulopathy of trauma shock or trauma-induced coagulopathy. The recognition of acute traumatic coagulopathy is important, because we now understand that its presence is a prognostic indicator, as it is associated with poor clinical outcome. This has driven a change in clinical management, so that the previous approach of maintaining an adequate circulating volume and oxygen carrying capacity before, as a secondary event, dealing with coagulopathy, has changed to haemostatic resuscitation as early as possible. While there is as yet no universally accepted assay or definition, many experts use prolongation of the prothrombin time to indicate that there is, indeed, a coagulopathy. Hypoxia, acidosis and hypothermia and hormonal, immunological and cytokine production, alongside consumption and blood loss, and the dilutional effects of resuscitation may occur to varying extents depending on the type of tissue damaged, the type and extent of injury, predisposing to, or amplifying, activation of coagulation, platelets, fibrinolysis. These are discussed in detail within the article.
Collapse
Affiliation(s)
- A Cap
- Uniformed Services University, Blood Research Program, US Army Institute of Surgical Research, Sam Houston, Texas, USA
| | | |
Collapse
|
48
|
Rousseau M, Belleannee C, Duchez AC, Cloutier N, Levesque T, Jacques F, Perron J, Nigrovic PA, Dieude M, Hebert MJ, Gelb MH, Boilard E. Detection and quantification of microparticles from different cellular lineages using flow cytometry. Evaluation of the impact of secreted phospholipase A2 on microparticle assessment. PLoS One 2015; 10:e0116812. [PMID: 25587983 PMCID: PMC4294685 DOI: 10.1371/journal.pone.0116812] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2014] [Accepted: 12/15/2014] [Indexed: 11/20/2022] Open
Abstract
Microparticles, also called microvesicles, are submicron extracellular vesicles produced by plasma membrane budding and shedding recognized as key actors in numerous physio(patho)logical processes. Since they can be released by virtually any cell lineages and are retrieved in biological fluids, microparticles appear as potent biomarkers. However, the small dimensions of microparticles and soluble factors present in body fluids can considerably impede their quantification. Here, flow cytometry with improved methodology for microparticle resolution was used to detect microparticles of human and mouse species generated from platelets, red blood cells, endothelial cells, apoptotic thymocytes and cells from the male reproductive tract. A family of soluble proteins, the secreted phospholipases A2 (sPLA2), comprises enzymes concomitantly expressed with microparticles in biological fluids and that catalyze the hydrolysis of membrane phospholipids. As sPLA2 can hydrolyze phosphatidylserine, a phospholipid frequently used to assess microparticles, and might even clear microparticles, we further considered the impact of relevant sPLA2 enzymes, sPLA2 group IIA, V and X, on microparticle quantification. We observed that if enriched in fluids, certain sPLA2 enzymes impair the quantification of microparticles depending on the species studied, the source of microparticles and the means of detection employed (surface phosphatidylserine or protein antigen detection). This study provides analytical considerations for appropriate interpretation of microparticle cytofluorometric measurements in biological samples containing sPLA2 enzymes.
Collapse
Affiliation(s)
- Matthieu Rousseau
- Centre de Recherche en Rhumatologie et Immunologie, Centre de Recherche du Centre Hospitalier Universitaire de Québec, Faculté de Médecine de l’Université Laval, Québec, QC, Canada
| | - Clemence Belleannee
- Centre de Recherche du CHUQ and Département d’Obstétrique-Gynécologie, Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Anne-Claire Duchez
- Centre de Recherche en Rhumatologie et Immunologie, Centre de Recherche du Centre Hospitalier Universitaire de Québec, Faculté de Médecine de l’Université Laval, Québec, QC, Canada
| | - Nathalie Cloutier
- Centre de Recherche en Rhumatologie et Immunologie, Centre de Recherche du Centre Hospitalier Universitaire de Québec, Faculté de Médecine de l’Université Laval, Québec, QC, Canada
| | - Tania Levesque
- Centre de Recherche en Rhumatologie et Immunologie, Centre de Recherche du Centre Hospitalier Universitaire de Québec, Faculté de Médecine de l’Université Laval, Québec, QC, Canada
| | | | - Jean Perron
- Centre Hospitalier Universitaire de Québec, Québec, Canada
| | - Peter A. Nigrovic
- Division of Rheumatology, Immunology and Allergy, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States of America
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Melanie Dieude
- Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Marie-Josee Hebert
- Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Michael H. Gelb
- Department of Chemistry, University of Washington, Seattle, WA, United States of America
| | - Eric Boilard
- Centre de Recherche en Rhumatologie et Immunologie, Centre de Recherche du Centre Hospitalier Universitaire de Québec, Faculté de Médecine de l’Université Laval, Québec, QC, Canada
- * E-mail:
| |
Collapse
|
49
|
|
50
|
Nekludov M, Mobarrez F, Gryth D, Bellander BM, Wallen H. Formation of microparticles in the injured brain of patients with severe isolated traumatic brain injury. J Neurotrauma 2014; 31:1927-33. [PMID: 24956150 DOI: 10.1089/neu.2013.3168] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The potential pathophysiological role of circulating microparticles (MPs) has been recognized in various conditions, such as cardiovascular and thrombotic diseases. Traumatic brain injury (TBI) has a complex pathophysiology that involves coagulopathy and inflammation. We investigated endothelial-, platelet-, and leukocyte-derived microparticles (EMPs, PMPs, and LMPs, respectively) in 16 patients with severe isolated TBI. Arterial and cerebrovenous samples were taken repeatedly, during 1-72 h after injury. Subpopulations of MPs, exposing tissue factor (TF) and P-selection, were also studied. MP counts in cerebrovenous samples, irrespective of cellular origin, were higher in TBI cases, compared to healthy controls (peak levels of EMPs were approximately 7 times higher, PMPs 1.4 times higher, and LMPs 2 times higher, respectively; p<0.001 for all). MP counts declined sharply from high levels shortly after the trauma toward slightly elevated levels 72 h later. EMPs and PMPs exposing TF, as well as PMPs exposing P-selection, showed a transcranial gradient with higher concentration in cerebrovenous, compared to arterial, samples. In contrast, LMPs exposing TF were higher in arterial samples, suggesting accumulation of LMPs in the brain. We conclude that the pattern of circulating MPs is altered after TBI. PMPs exposing P-selection and EMPs exposing TF seem to be generated in the injured brain, whereas LMPs exposing TF are accumulated. The pathophysiological significance of these changes in MP pattern in TBI should be further investigated. Including MPs exposing brain-specific antigens in the assessment of brain injury would give further information of origin and likely give additional information of the size of the injury, given that the MP phenotypes investigated in the present study are not brain-specific markers.
Collapse
Affiliation(s)
- Michael Nekludov
- 1 Karolinska Institute, Department of Physiology and Pharmacology, Section for Anesthesiology, Karolinska University Hospital , Stockholm, Sweden
| | | | | | | | | |
Collapse
|