101
|
Effect of Coagulation Factor Concentrates on Markers of Endothelial Cell Damage in Experimental Hemorrhagic Shock. Shock 2020; 52:497-505. [PMID: 30407369 DOI: 10.1097/shk.0000000000001286] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Plasma-based resuscitation showed protective effects on the endothelial glycocalyx compared with crystalloid resuscitation. There is paucity of data regarding the effect of coagulation factor concentrates (CFC) on the glycocalyx in hemorrhagic shock (HS). We hypothesized that colloid-based resuscitation supplemented with CFCs offers a therapeutic value to treat endothelial damage following HS. METHODS Eighty-four rats were subjected to pressure-controlled (mean arterial pressure (MAP) 30-35 mm Hg) and lab-guided (targeted cutoff: lactate >2.2. mmol/L and base deficit > 5.5 mmol/L) HS. Animals were resuscitated with fresh frozen plasma (FFP), human albumin (HA) or Ringer's lactate (RL) and RL or HA supplemented with fibrinogen concentrate (FC) or prothrombin complex concentrate (PCC). Serum epinephrine and the following markers of endothelial damage were assessed at baseline and at the end-of-observation (120 min after shock was terminated): syndecan-1, heparan sulfate, and soluble vascular endothelial growth factor receptor 1 (sVEGFR 1). RESULTS Resuscitation with FFP had no effect on sVEGFR1 compared with crystalloid-based resuscitation (FFP: 19.3 ng/mL vs. RL: 15.9 ng/mL; RL+FC: 19.7 ng/mL; RL+PCC: 18.9 ng/mL; n.s.). At the end-of-observation, syndecan-1 was similar among all groups. Interestingly, HA+FC treated animals displayed the highest syndecan-1 concentration (12.07 ng/mL). Resuscitation with FFP restored heparan sulfate back to baseline (baseline: 36 ng/mL vs. end-of-observation: 36 ng/mL). CONCLUSION The current study revealed that plasma-based resuscitation normalized circulating heparan sulfate but not syndecan-1. Co-administration of CFC had no further effect on glycocalyx shedding suggesting a lack of its therapeutic potential. LEVEL OF EVIDENCE VExperimental in vivo study.
Collapse
|
102
|
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
|
103
|
Is there a place for regional anesthesia in nonoperating room anesthesia? Curr Opin Anaesthesiol 2020; 33:561-565. [DOI: 10.1097/aco.0000000000000897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
104
|
Li K, Wu H, Pan F, Chen L, Feng C, Liu Y, Hui H, Cai X, Che H, Ma Y, Li T. A Machine Learning-Based Model to Predict Acute Traumatic Coagulopathy in Trauma Patients Upon Emergency Hospitalization. Clin Appl Thromb Hemost 2020; 26:1076029619897827. [PMID: 31908189 PMCID: PMC7098202 DOI: 10.1177/1076029619897827] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Acute traumatic coagulopathy (ATC) is an extremely common but silent murderer; this condition presents early after trauma and impacts approximately 30% of severely injured patients who are admitted to emergency departments (EDs). Given that conventional coagulation indicators usually require more than 1 hour after admission to yield results—a limitation that frequently prevents the ability for clinicians to make appropriate interventions during the optimal therapeutic window—it is clearly of vital importance to develop prediction models that can rapidly identify ATC; such models would also facilitate ancillary resource management and clinical decision support. Using the critical care Emergency Rescue Database and further collected data in ED, a total of 1385 patients were analyzed and cases with initial international normalized ratio (INR) values >1.5 upon admission to the ED met the defined diagnostic criteria for ATC; nontraumatic conditions with potentially disordered coagulation systems were excluded. A total of 818 individuals were collected from Emergency Rescue Database as derivation cohorts, then were split 7:3 into training and test data sets. A Pearson correlation matrix was used to initially identify likely key clinical features associated with ATC, and analysis of data distributions was undertaken prior to the selection of suitable modeling tools. Both machine learning (random forest) and traditional logistic regression were deployed for prediction modeling of ATC. After the model was built, another 587 patients were further collected in ED as validation cohorts. The ATC prediction models incorporated red blood cell count, Shock Index, base excess, lactate, diastolic blood pressure, and potential of hydrogen. Of 818 trauma patients filtered from the database, 747 (91.3%) patients did not present ATC (INR ≤ 1.5) and 71 (8.7%) patients had ATC (INR > 1.5) upon admission to the ED. Compared to the logistic regression model, the model based on the random forest algorithm showed better accuracy (94.0%, 95% confidence interval [CI]: 0.922-0.954 to 93.5%, 95% CI: 0.916-0.95), precision (93.3%, 95% CI: 0.914-0.948 to 93.1%, 95% CI: 0.912-0.946), F1 score (93.4%, 95% CI: 0.915-0.949 to 92%, 95% CI: 0.9-0.937), and recall score (94.0%, 95% CI: 0.922-0.954 to 93.5%, 95% CI: 0.916-0.95) but yielded lower area under the receiver operating characteristic curve (AU-ROC) (0.810, 95% CI: 0.673-0.918 to 0.849, 95% CI: 0.732-0.944) for predicting ATC in the trauma patients. The result is similar in the validation cohort. The values for classification accuracy, precision, F1 score, and recall score of random forest model were 0.916, 0.907, 0.901, and 0.917, while the AU-ROC was 0.830. The values for classification accuracy, precision, F1 score, and recall score of logistic regression model were 0.905, 0.887, 0.883, and 0.905, while the AU-ROC was 0.858. We developed and validated a prediction model based on objective and rapidly accessible clinical data that very confidently identify trauma patients at risk for ATC upon their arrival to the ED. Beyond highlighting the value of ED initial laboratory tests and vital signs when used in combination with data analysis and modeling, our study illustrates a practical method that should greatly facilitates both warning and guided target intervention for ATC.
Collapse
Affiliation(s)
- Kaiyuan Li
- Department of Emergency, The First Medical Center to Chinese People's Liberation Army General Hospital, Beijing, China
| | - Huitao Wu
- National Engineering Laboratory for Medical Big Data Application Technology, The First Medical Center to Chinese People's Liberation Army General Hospital, Beijing, China
| | - Fei Pan
- Department of Emergency, The First Medical Center to Chinese People's Liberation Army General Hospital, Beijing, China
| | - Li Chen
- Department of Emergency, The First Medical Center to Chinese People's Liberation Army General Hospital, Beijing, China
| | - Cong Feng
- Department of Emergency, The First Medical Center to Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yihao Liu
- Department of Emergency, The First Medical Center to Chinese People's Liberation Army General Hospital, Beijing, China
| | - Hui Hui
- Department of Emergency, The First Medical Center to Chinese People's Liberation Army General Hospital, Beijing, China
| | - Xiaoyu Cai
- Department of Blood Transfusion, The First Medical Center to Chinese People's Liberation Army General Hospital, Beijing, China
| | - Hebin Che
- National Engineering Laboratory for Medical Big Data Application Technology, The First Medical Center to Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yulong Ma
- Anesthesia and Operation Center, The First Medical Center to Chinese PLA General Hospital, Beijing, China
| | - Tanshi Li
- Department of Emergency, The First Medical Center to Chinese People's Liberation Army General Hospital, Beijing, China
| |
Collapse
|
105
|
Leal-Noval SR, Fernández Pacheco J, Casado Méndez M, Cuenca-Apolo D, Múñoz-Gómez M. Current perspective on fibrinogen concentrate in critical bleeding. Expert Rev Clin Pharmacol 2020; 13:761-778. [PMID: 32479129 DOI: 10.1080/17512433.2020.1776608] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION . Massive hemorrhage continues to be a treatable cause of death. Its management varies from prefixed ratio-driven administration of blood components to goal-directed therapy based on point-of-care testing and administration of coagulation factor concentrates. AREAS COVERED . We review the current role of fibrinogen concentrate (FC) for the management of massive hemorrhage, either administered without coagulation testing in life-threatening hemorrhage, or within an algorithm based on viscoelastic hemostatic assays and plasma fibrinogen level. We identified relevant guidelines, meta-analyzes, randomized controlled trials, and observational studies that included indications, dosage, and adverse effects of FC, especially thromboembolic events. EXPERT OPINION . Moderate- to high-grade evidence supports the use of FC for the treatment of severe hemorrhage in trauma and cardiac surgery; a lower grade of evidence is available for its use in postpartum hemorrhage and end-stage liver disease. Pre-emptive FC administration in non-bleeding patients is not recommended. FC should be administered early, in a goal-directed manner, guided by early amplitude of clot firmness parameters (A5- or A10-FIBTEM) or hypofibrinogenemia. Further investigation is required into the early use of FC, as well as its potential advantages over cryoprecipitate, and whether or not its administration at high doses leads to a greater risk of adverse events.
Collapse
Affiliation(s)
- Santiago R Leal-Noval
- Neuro Critical Care Department, University Hospital "Virgen Del Rocío" and Institute of Biomedicine "IBIS" , 41013, Seville, Spain
| | - Jose Fernández Pacheco
- Pharmacy and Statistics and Design, University Hospital "Virgen Del Rocío" and Institute of Biomedicine "IBIS" , 41013, Seville, Spain
| | - Manuel Casado Méndez
- Critical Care Department, University Hospital "Virgen Del Rocío" and Institute of Biomedicine "IBIS" , 41013, Seville, Spain
| | - Diego Cuenca-Apolo
- Critical Care Department, University Hospital "Virgen Del Rocío" and Institute of Biomedicine "IBIS" , 41013, Seville, Spain
| | - Manuel Múñoz-Gómez
- Department of Surgical Specialties, Biochemistry and Immunology, University of Málaga , 29071, Málaga, Spain
| |
Collapse
|
106
|
Maegele M. The Diagnosis and Treatment of Acute Traumatic Bleeding and Coagulopathy. DEUTSCHES ARZTEBLATT INTERNATIONAL 2020; 116:799-806. [PMID: 31847951 DOI: 10.3238/arztebl.2019.0799] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 06/13/2019] [Accepted: 09/06/2019] [Indexed: 11/27/2022]
Abstract
BACKGROUND Uncontrolled bleeding with trauma-induced coagulopathy (TIC) is still the most common avoidable cause of death in multiple trauma. The aging of the population has led to an increasing number of bleeding trauma patients with pre-existing anticoagulation. Such patients are not treated uniformly, even in major trauma centers. METHODS This review is based on a selective search of the literature (Medline/PubMed, Cochrane Reviews) and summarizes current treatment recommendations, including those of the newly revised European trauma guidelines. RESULTS The treatment of traumatic hemorrhage begins at the site of the accident, with compression, tourniquets, pelvic binders, and rapid transport to a certified trauma center. The early use of tourniquets was shown to lessen the trans- fusion requirement (packed red blood cells: 2.0 ± 0.1 vs. 9.3 ± 0.6; p < 0.001; fresh frozen plasma concentrates: 1.4 ± 0.08 vs. 6.2 ± 0.4; p < 0.001), while external pelvic stabilization was shown to reduce mortality (19.1% vs. 33.3%). Upon the patient's arrival in the hospital, steps are taken to measure, monitor, and support clotting function. Bleeding is controlled surgically according to the principles of damage control. Modern clotting management consists of goal-oriented, individualized therapy, including the use of point-of-care viscoelastic test procedures. Idarucizumab can be used as an antidote to the thrombin inhibitor dabigatran, andexanet alpha as an antidote to factor Xa inhibitors. CONCLUSION The evidence-based treatment of patients with hemorrhage from severe trauma, in accordance with the existing guidelines, can improve the clinical outcome. Corresponding algorithms, adapted to local logistics and infrastructure, must be developed and implemented.
Collapse
Affiliation(s)
- Marc Maegele
- Department of Orthopedic Surgery, Trauma Surgery, and Sports Medicine, Cologne Merheim Medical Center, Witten/Herdecke University, Cologne
| |
Collapse
|
107
|
Li B, Zhou X, Yi TL, Xu ZW, Peng DW, Guo Y, Guo YM, Cao YL, Zhu L, Zhang S, Cheng SX. Bloodletting Puncture at Hand Twelve Jing-Well Points Improves Neurological Recovery by Ameliorating Acute Traumatic Brain Injury-Induced Coagulopathy in Mice. Front Neurosci 2020; 14:403. [PMID: 32581664 PMCID: PMC7290011 DOI: 10.3389/fnins.2020.00403] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 04/02/2020] [Indexed: 12/14/2022] Open
Abstract
Traumatic brain injury (TBI) contributes to hypocoagulopathy associated with prolonged bleeding and hemorrhagic progression. Bloodletting puncture therapy at hand twelve Jing-well points (BL-HTWP) has been applied as a first aid measure in various emergent neurological diseases, but the detailed mechanisms of the modulation between the central nervous system and systemic circulation after acute TBI in rodents remain unclear. To investigate whether BL-HTWP stimulation modulates hypocoagulable state and exerts neuroprotective effect, experimental TBI model of mice was produced by the controlled cortical impactor (CCI), and treatment with BL-HTWP was immediately made after CCI. Then, the effects of BL-HTWP on the neurological function, cerebral perfusion state, coagulable state, and cerebrovascular histopathology post-acute TBI were determined, respectively. Results showed that BL-HTWP treatment attenuated cerebral hypoperfusion and improve neurological recovery post-acute TBI. Furthermore, BL-HTWP stimulation reversed acute TBI-induced hypocoagulable state, reduced vasogenic edema and cytotoxic edema by regulating multiple hallmarks of coagulopathy in TBI. Therefore, we conclude for the first time that hypocoagulopathic state occurs after acute experimental TBI, and the neuroprotective effect of BL-HTWP relies on, at least in part, the modulation of hypocoagulable state. BL-HTWP therapy may be a promising strategy for acute severe TBI in the future.
Collapse
Affiliation(s)
- Bo Li
- Tianjin Key Laboratory of Neurotrauma Repair, Institute of Neurotrauma Repair of Characteristic Medical Center of Chinese People's Armed Police Force (PAP), Tianjin, China.,Acupuncture Research Center, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiu Zhou
- Tianjin Key Laboratory of Neurotrauma Repair, Institute of Neurotrauma Repair of Characteristic Medical Center of Chinese People's Armed Police Force (PAP), Tianjin, China
| | - Tai-Long Yi
- Tianjin Key Laboratory of Neurotrauma Repair, Institute of Neurotrauma Repair of Characteristic Medical Center of Chinese People's Armed Police Force (PAP), Tianjin, China
| | - Zhong-Wei Xu
- Central Laboratory of Logistics University of Chinese People's Armed Police Force (PAP), Tianjin, China
| | - Ding-Wei Peng
- Tianjin Key Laboratory of Neurotrauma Repair, Institute of Neurotrauma Repair of Characteristic Medical Center of Chinese People's Armed Police Force (PAP), Tianjin, China
| | - Yi Guo
- Acupuncture Research Center, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yong-Ming Guo
- Acupuncture Research Center, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yu-Lin Cao
- Zhenxigu Medical Research Center, Beijing, China
| | - Lei Zhu
- Department of Spine Surgery, Xi'an Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Sai Zhang
- Tianjin Key Laboratory of Neurotrauma Repair, Institute of Neurotrauma Repair of Characteristic Medical Center of Chinese People's Armed Police Force (PAP), Tianjin, China
| | - Shi-Xiang Cheng
- Tianjin Key Laboratory of Neurotrauma Repair, Institute of Neurotrauma Repair of Characteristic Medical Center of Chinese People's Armed Police Force (PAP), Tianjin, China
| |
Collapse
|
108
|
James V, Chong SL, Shetty SS, Ong GY. Early coagulopathy in children with isolated blunt head injury is associated with mortality and poor neurological outcomes. J Neurosurg Pediatr 2020; 25:663-669. [PMID: 32114542 DOI: 10.3171/2019.12.peds19531] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 12/30/2019] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Traumatic brain injury (TBI) is the leading cause of long-term disability and death in children and adolescents globally. Long-term adverse outcomes, including physical, cognitive, and behavioral sequelae, have been reported after TBI in a significant number of pediatric patients. In this study the authors sought to investigate the epidemiology of TBI-associated coagulopathy and its association with mortality and poor neurological outcome in a pediatric population with isolated moderate to severe blunt head injury treated at the authors' institution. METHODS This retrospective study was conducted in the children's emergency department between January 2010 and December 2016. Children < 18 years old who presented with isolated moderate to severe blunt head injury were included in the study. The authors collected data on patient demographics, clinical presentation, and TBI management. Outcomes studied were death and poor neurological outcome defined by a score of < 7 (death, moderate to severe neurological disability) at 6 months postinjury on the pediatric version of the Glasgow Outcome Scale-Extended (GOS-E Peds). RESULTS In 155 pediatric patients who presented with isolated moderate to severe blunt head injury, early coagulopathy was observed in 33 (21.3%) patients during the initial blood investigations done in the emergency department. The mean (SD) age of the study group was 7.03 (5.08) years and the predominant mechanism of injury was fall from height (65.2%). The median Abbreviated Injury Scale of the head (AIS head) score was 4 and the median GCS score was 13 (IQR 12-15). TBI-associated coagulopathy was independently associated with GOS-E Peds score < 7 (p = 0.02, adjusted OR 6.07, 95% CI 1.32-27.83). The overall mortality rate was 5.8%. After adjusting for confounders, only AIS head score and hypotension at triage remained significantly associated with TBI-associated coagulopathy. CONCLUSIONS TBI-associated coagulopathy was independently associated with GOS-E Peds score < 7 at 6 months postinjury. Larger prospective studies are needed to investigate the use of TBI-associated coagulopathy to prognosticate these critical clinical outcomes.
Collapse
|
109
|
Huang S, Tang Y, Lin W, Wen K, Han W, Lin Z, Han M. Study on coagulation profiles and platelet function in trauma-induced coagulopathy caused by three types of injury. Injury 2020; 51:1312-1320. [PMID: 32093941 DOI: 10.1016/j.injury.2020.02.081] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Accepted: 02/16/2020] [Indexed: 02/05/2023]
Abstract
BACKGROUND Traumatic coagulopathy is a major public health issue globally with undefined mechanisms. We established rat models of hemorrhagic shock (HS), multiple injury (MI) and traumatic brain injury (TBI) to investigate the diversity of traumatic coagulopathy, especially platelet dysfunction. METHODS Seventy male SD rats were divided randomly into seven groups(n = 10): control, HS30min, HS3h, MI30min, MI3h, TBI30min and TBI3h. Plasma or whole blood was collected for conventional coagulation tests, thromboelastography and platelet mapping. X-ray, 7T magnetic resonance imaging and hematoxylin-eosin staining of injured tissues were conducted to confirm the injuries of rats model. RESULTS The activated partial thromboplastin time (aPTT) prolonged significantly in HS30min and MI3h groups, compared with those in control (P = 0.0403 and P = 0.0076, respectively). R values decreased in HS30min and HS3h groups, compared with those in control (P < 0.0001 and P < 0.0001, respectively). The maximum amplitude (MA) were 71.8 ± 0.6 mm, 71.9 ± 0.5 mm, 71.8 ± 0.7 mm, 70.0 ± 0.7 mm, 72.6 ± 0.9 mm, 70.4 ± 0.9 mm in HS30min, HS3h, MI30min, MI3h, TBI30min and TBI3h groups respectively, which were lower than those in control (P = 0.0304, P = 0.0205, P = 0.0431, P = 0.0007 and P = 0.0066, respectively). The platelet counts were 539±46 × 109/L, 523±31 × 109/L, 629 ± 18 × 109/L and 636±20 × 109/L in HS30min, HS3h, MI3h and TBI3h groups respectively, which were lower than those in control (P = 0.0040, P = 0.0001, P = 0.0127 and P = 0.0232, respectively). The adenosine diphosphate (ADP) inhibition rate decreased in HS30min group, compared with that in control (P = 0.0355). While, ADP inhibition rate increased in HS3h and TBI3h groups (P = 0.0041 and P = 0.0433 vs. control, respectively). The arachidonic acid (AA) inhibition rate increased in MI30min and MI3h groups, compared with control (P = 0.0029 and P = 0.0185, respectively). CONCLUSION These results demonstrated that it might be the failure of forming a strong clot instead of the prolonged clot time, which contributed to traumatic coagulopathy. The platelet dysfunctions might contribute to trauma-induced coagulopathy in different ways. The loss of platelets might be the main reason for HS-induced coagulopathy. While, AA-dependent pathway inhibition might account for MI-induced coagulopathy. ADP-dependent pathway inhibition might be the major contributor for TBI-induced coagulopathy.
Collapse
Affiliation(s)
- Sunhua Huang
- The Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Yao Tang
- The Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Wenhao Lin
- The Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Ke Wen
- Department of Microsurgery, Taihe Hospital, Shiyan, China
| | - Wei Han
- Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen, China
| | - Zhexuan Lin
- Bio-analytical Laboratory, Shantou University Medical College, Shantou, China.
| | - Ming Han
- Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen, China.
| |
Collapse
|
110
|
Alvikas J, Myers SP, Wessel CB, Okonkwo DO, Joseph B, Pelaez C, Dosberstein C, Guillotte AR, Rosengart MR, Neal MD. A systematic review and meta-analysis of traumatic intracranial hemorrhage in patients taking prehospital antiplatelet therapy: Is there a role for platelet transfusions? J Trauma Acute Care Surg 2020; 88:847-854. [PMID: 32118818 PMCID: PMC7431190 DOI: 10.1097/ta.0000000000002640] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
BACKGROUND Platelet transfusion has been utilized to reverse platelet dysfunction in patients on preinjury antiplatelets who have sustained a traumatic intracranial hemorrhage (tICH); however, there is little evidence to substantiate this practice. The objective of this study was to perform a systematic review on the impact of platelet transfusion on survival, hemorrhage progression and need for neurosurgical intervention in patients with tICH on prehospital antiplatelet medication. METHODS Controlled, observational and randomized, prospective and retrospective studies describing tICH, preinjury antiplatelet use, and platelet transfusion reported in PubMed, Embase, Cochrane Reviews, Cochrane Trials and Cochrane DARE databases between January 1987 and March 2019 were included. Investigations of concomitant anticoagulant use were excluded. Risk of bias was assessed using the Newcastle-Ottawa scale. We calculated pooled estimates of relative effect of platelet transfusion on the risk of death, hemorrhage progression and need for neurosurgical intervention using the methods of Dersimonian-Laird random-effects meta-analysis. Sensitivity analysis established whether study size contributed to heterogeneity. Subgroup analyses determined whether antiplatelet type, additional blood products/reversal agents, or platelet function assays impacted effect size using meta-regression. RESULTS Twelve of 18,609 screened references were applicable to our questions and were qualitatively and quantitatively analyzed. We found no association between platelet transfusion and the risk of death in patients with tICH taking prehospital antiplatelets (odds ratio [OR], 1.29; 95% confidence interval [CI], 0.76-2.18; p = 0.346; I = 32.5%). There was no significant reduction in hemorrhage progression (OR, 0.88; 95% CI, 0.34-2.28; p = 0.788; I = 78.1%). There was no significant reduction in the need for neurosurgical intervention (OR, 1.00; 95% CI, 0.53-1.90, p = 0.996; I = 59.1%; p = 0.032). CONCLUSION Current evidence does not support the use of platelet transfusion in patients with tICH on prehospital antiplatelets, highlighting the need for a prospective evaluation of this practice. LEVEL OF EVIDENCE Systematic Reviews and Meta-Analyses, Level III.
Collapse
Affiliation(s)
- Jurgis Alvikas
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Sara P. Myers
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Charles B. Wessel
- University of Pittsburgh Health Sciences Library System, Pittsburgh, PA
| | - David O. Okonkwo
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Bellal Joseph
- Department of Surgery, University of Arizona, Tucson, AZ
| | | | - Cody Dosberstein
- Warren Alpert Medical School of Brown University, Providence, RI
| | - Andrew R. Guillotte
- Division of Neurological Surgery, University of Missouri School of Medicine, Columbia, MO
| | - Matthew R. Rosengart
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA
- Department of Critical Care Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Matthew D. Neal
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA
- Department of Critical Care Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA
| |
Collapse
|
111
|
Platelet dysfunction during trauma involves diverse signaling pathways and an inhibitory activity in patient-derived plasma. J Trauma Acute Care Surg 2020; 86:250-259. [PMID: 30531331 DOI: 10.1097/ta.0000000000002140] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Trauma-induced coagulopathy occurs in about 25% of injured patients and accounts for about 10% of deaths worldwide. Upon injury, hemostatic function may decline due to vascular dysfunction, clotting factor deficiencies, hyperfibrinolysis, and/or platelet dysfunction. We investigated agonist-induced calcium signaling in platelets obtained over time from trauma patients. METHODS Platelets from trauma patients and healthy donors were monitored via intracellular calcium mobilization and flow cytometry markers (α2bβ3 activation, P-selectin display, and phosphatidylserine exposure) following stimulation with a panel of agonists (adenosine 5'-diphosphate sodium salt, U46619, convulxin, PAR-1/4 activating peptides, iloprost) used in isolation or in pairwise tests. Furthermore, healthy donor platelets were tested in heterologous plasma isolated from healthy subjects and trauma patients. RESULTS When exposed to agonists over the first 24 hours postinjury, trauma patient platelets mobilized less calcium in comparison to healthy platelets. Partial recovery of platelet activity was observed in about a third of patients after 120 hours, although not fully obtaining healthy baseline function. Flow cytometry markers of trauma platelets were similar to healthy platelets prior to stimulation, but were depressed in trauma platelets stimulated with adenosine 5'-diphosphate sodium salt or convulxin. Also, washed healthy platelets showed a significant reduction in calcium mobilization when reconstituted in plasma from trauma patients, relative to healthy plasma, at all plasma doses tested. CONCLUSION Platelet dysfunction in trauma patients included poor response to multiple agonists relevant to hemostatic function. Furthermore, the inhibitor effect of patient plasma on healthy platelets suggests that soluble plasma species may downregulate endogenous or transfused platelets during trauma.
Collapse
|
112
|
Dyer MR, Plautz WE, Ragni MV, Alexander W, Haldeman S, Sperry JL, Guyette FX, Zuckerbraun BS, Rollins-Raval MA, Raval JS, Neal MD. Traumatic injury results in prolonged circulation of ultralarge von Willebrand factor and a reduction in ADAMTS13 activity. Transfusion 2020; 60:1308-1318. [PMID: 32441353 DOI: 10.1111/trf.15856] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 02/19/2020] [Accepted: 04/05/2020] [Indexed: 01/02/2023]
Abstract
BACKGROUND Increases in plasma von Willebrand Factor (VWF) levels, accompanied by decreases in the metalloprotease ADAMTS13, have been demonstrated soon after traumatic injury while downstream effects remain unclear. STUDY DESIGN AND METHODS A cohort of 37 injured trauma patients from a randomized control trial investigating the use of prehospital plasma transfusion were analyzed for activity and antigen levels of ADAMTS13 and VWF at 0 and 24 hours after admission. Relevant clinical data were abstracted from the medical records. Trauma patient plasma was analyzed via agarose gel electrophoresis to evaluate the effects of injury on VWF multimer composition compared to healthy controls. RESULTS von Willebrand factor levels were elevated at presentation (189% [110%-263%] vs. 95% [74%-120%]), persisting through 24 hours (213% [146%-257%] vs. 132% [57%-160%]), compared to healthy controls. Ultralarge VWF (UL-VWF) forms were elevated in trauma patients at both 0 and 24 hours, when compared to pooled normal plasma (10.0% [8.9%-14.3%] and 11.3% [9.1%-21.2%], respectively, vs. 0.6%). Circulating plasma ADAMTS13 activity was decreased at 0 hours (66% [47%-86%] vs. 100% [98%-100%]) and at 24 hours (72.5% [56%-87.3%] vs. 103% [103%-103%]) in trauma patients. ADAMTS13 activity independently predicted the development of coagulopathy and correlated with international normalized ratio, thromboelastography values, injury severity, and blood product transfusion. CONCLUSION Traumatic injury is associated with acute coagulopathy that is characterized by increased UL-VWF multimers and reduction in ADAMTS13, which correlates with blood loss, transfusion requirement, and injury severity. These findings suggest the potential for future trials targeting ADAMTS13 repletion to enhance clearance of VWF multimers.
Collapse
Affiliation(s)
- Mitchell R Dyer
- Pittsburgh Trauma Research Center and the Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - William E Plautz
- University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Margaret V Ragni
- Division of Hematology/Oncology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Wyeth Alexander
- University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Shannon Haldeman
- Pittsburgh Trauma Research Center and the Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jason L Sperry
- Pittsburgh Trauma Research Center and the Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Francis X Guyette
- Department of Emergency Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Brian S Zuckerbraun
- Pittsburgh Trauma Research Center and the Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Marian A Rollins-Raval
- Department of Pathology, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina, USA.,Department of Pathology, University of New Mexico, Albuquerque, New Mexico, USA
| | - Jay S Raval
- Department of Pathology, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina, USA.,Department of Pathology, University of New Mexico, Albuquerque, New Mexico, USA
| | - Matthew D Neal
- Pittsburgh Trauma Research Center and the Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | |
Collapse
|
113
|
Mitrophanov AY, Szlam F, Sniecinski RM, Levy JH, Reifman J. Controlled Multifactorial Coagulopathy: Effects of Dilution, Hypothermia, and Acidosis on Thrombin Generation In Vitro. Anesth Analg 2020; 130:1063-1076. [PMID: 31609256 DOI: 10.1213/ane.0000000000004479] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Coagulopathy and hemostatic abnormalities remain a challenge in patients following trauma and major surgery. Coagulopathy in this setting has a multifactorial nature due to tissue injury, hemodilution, hypothermia, and acidosis, the severity of which may vary. In this study, we combined computational kinetic modeling and in vitro experimentation to investigate the effects of multifactorial coagulopathy on thrombin, the central enzyme in the coagulation system. METHODS We measured thrombin generation in platelet-poor plasma from 10 healthy volunteers using the calibrated automated thrombogram assay (CAT). We considered 3 temperature levels (31°C, 34°C, and 37°C), 3 pH levels (6.9, 7.1, and 7.4), and 3 degrees of dilution with normal saline (no dilution, 3-fold dilution, and 5-fold dilution). We measured thrombin-generation time courses for all possible combinations of these conditions. For each combination, we analyzed 2 scenarios: without and with (15 nM) supplementation of thrombomodulin, a key natural regulator of thrombin generation. For each measured thrombin time course, we recorded 5 quantitative parameters and analyzed them using multivariable regression. Moreover, for multiple combinations of coagulopathic conditions, we performed routine coagulation tests: prothrombin time (PT) and activated partial thromboplastin time (aPTT). We compared the experimental results with simulations using a newly developed version of our computational kinetic model of blood coagulation. RESULTS Regression analysis allowed us to identify trends in our data (P < 10). In both model simulations and experiments, dilution progressively reduced the peak of thrombin generation. However, we did not experimentally detect the model-predicted delay in the onset of thrombin generation. In accord with the model predictions, hypothermia delayed the onset of thrombin generation; it also increased the thrombin peak time (up to 1.30-fold). Moreover, as predicted by the kinetic model, the experiments showed that hypothermia increased the area under the thrombin curve (up to 1.97-fold); it also increased the height of the thrombin peak (up to 1.48-fold). Progressive acidosis reduced the velocity index by up to 24%; acidosis-induced changes in other thrombin generation parameters were much smaller or none. Acidosis increased PT by 14% but did not influence aPTT. In contrast, dilution markedly prolonged both PT and aPTT. In our experiments, thrombomodulin affected thrombin-generation parameters mainly in undiluted plasma. CONCLUSIONS Dilution with normal saline reduced the amount of generated thrombin, whereas hypothermia increased it and delayed the time of thrombin accumulation. In contrast, acidosis in vitro had little effect on thrombin generation.
Collapse
Affiliation(s)
- Alexander Y Mitrophanov
- From the The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, Maryland.,DoD Biotechnology High Performance Computing Software Applications Institute (BHSAI), Telemedicine and Advanced Technology Research Center, US Army Medical Research and Development Command, Ft Detrick, Maryland
| | - Fania Szlam
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, Georgia
| | - Roman M Sniecinski
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, Georgia
| | - Jerrold H Levy
- Departments of Anesthesiology, Critical Care, and Surgery, Duke University School of Medicine, Durham, North Carolina
| | - Jaques Reifman
- DoD Biotechnology High Performance Computing Software Applications Institute (BHSAI), Telemedicine and Advanced Technology Research Center, US Army Medical Research and Development Command, Ft Detrick, Maryland
| |
Collapse
|
114
|
Platelet factor 4 and β-thromboglobulin mRNAs in circulating microparticles of trauma patients as diagnostic markers for deep vein thrombosis. J Thromb Thrombolysis 2020; 50:525-532. [PMID: 32347511 DOI: 10.1007/s11239-020-02124-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Deep vein thrombosis (DVT) is a common complication after trauma. The development of markers to predict DVT in trauma patients is needed, and circulating microparticles (MPs) and their contents are possible candidates. In this study, we aimed to identify platelet factor 4 (PF4) and β-thromboglobulin (β-TG) mRNAs in circulating MPs as potential markers for DVT diagnosis in trauma patients. Fifteen trauma patients diagnosed with DVT and fifteen matched patients without DVT were included in this study. Fifteen healthy volunteers also were included as controls. Circulating MPs were obtained from the plasma of all study subjects. Annexin V+ MPs and platelet-derived MPs (PMPs) were quantified using flow cytometry. PF4 and β-TG mRNAs in MPs were determined by qPCR, and the common logarithm of relative quantitation (RQ) was calculated using the comparative Ct method. Receiver-operating characteristic (ROC) curves were performed to analyze the diagnostic value of PF4 and β-TG mRNAs. No significant differences were found in Annexin V+ MPs and PMPs levels between trauma patients with and without DVT. However, both PF4 and β-TG mRNAs in MPs from the DVT group were significantly higher than the non-DVT group and healthy controls (P = 0.014 for PF4, P = 0.010 for β-TG). The ROC curve analysis showed that both the PF4 mRNA (area-under curve (AUC) 0.756, P = 0.017) and the β-TG mRNA (AUC 0.751, P = 0.019) had a positive predictive value for DVT. This finding indicates that the PF4 and β-TG mRNAs in MPs may be used as potential biomarkers for DVT diagnosis in trauma patients.
Collapse
|
115
|
Moore HB, Gando S, Iba T, Kim PY, Yeh CH, Brohi K, Hunt BJ, Levy JH, Draxler DF, Stanworth S, Görlinger K, Neal MD, Schreiber MA, Barrett CD, Medcalf RL, Moore EE, Mutch NJ, Thachil J, Urano T, Thomas S, Scărlătescu E, Walsh M. Defining trauma-induced coagulopathy with respect to future implications for patient management: Communication from the SSC of the ISTH. J Thromb Haemost 2020; 18:740-747. [PMID: 32112533 DOI: 10.1111/jth.14690] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 11/12/2019] [Accepted: 11/25/2019] [Indexed: 12/22/2022]
Affiliation(s)
- Hunter B Moore
- Department of Surgery, University of Colorado, Denver, CO, USA
| | - Satoshi Gando
- Division of Acute and Critical Care Medicine, Department of Anesthesiology and Critical Care Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
- Department of Acute and Critical Care Medicine, Sapporo Higashi Tokushukai Hospital, Sapporo, Japan
| | - Toshiaki Iba
- Department of Emergency and Disaster Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Paul Y Kim
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- Thrombosis and Atherosclerosis Research Institute, Hamilton, ON, Canada
| | - Calvin H Yeh
- Department of Medicine, Division of Emergency Medicine, University of Toronto, Toronto, ON,, Canada
| | - Karim Brohi
- Queen Mary University of London, London, UK
- Centre for Trauma Sciences, London, UK
| | | | - Jerrold H Levy
- Department of Anesthesiology, Critical Care, and Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Dominik F Draxler
- Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria,, Australia
| | - Simon Stanworth
- Transfusion Medicine, NHS Blood and Transplant, Oxford, UK
- Department of Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- Radcliffe Department of Medicine, NIHR Oxford Biomedical Research Centre,, University of Oxford,, Oxford,, UK
| | - Klaus Görlinger
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Essen, Essen, Germany
- TEM Innovations GmbH, Munich, Germany
| | - Matthew D Neal
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Martin A Schreiber
- Department of Surgery, Oregon Health & Science University, Portland, OR, USA
| | - Christopher D Barrett
- Koch Institute for Integrative Cancer Research, Center for Precision Cancer Medicine, Massachusetts Institute of Technology, Cambridge, MA, USA
- Division of Acute Care Surgery and Critical Care, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Robert L Medcalf
- Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria,, Australia
| | - Ernest E Moore
- Ernest E. Moore Shock Trauma Center at Denver Health, University of Colorado, Denver, CO, USA
| | - Nicola J Mutch
- Aberdeen Cardiovascular and Diabetes Centre, School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Jecko Thachil
- Department of Haematology, Manchester Royal Infirmary, Manchester, UK
| | - Tetsumei Urano
- Department of Medical Physiology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Scott Thomas
- Beacon Medical Group Trauma and Surgical Research Services, South Bend, IN, USA
| | - Ecaterina Scărlătescu
- Department of Anaesthesia and Intensive Care, Fundeni Clinical Institute, Bucharest, Romania
| | - Mark Walsh
- Beacon Medical Group Trauma and Surgical Research Services, South Bend, IN, USA
- Departments of Emergency and Internal Medicine, Saint Joseph Regional Medical Center, Mishawaka, IN, USA
- Indiana University School of Medicine, South Bend Campus, South Bend, IN, USA
| |
Collapse
|
116
|
Fletcher-Sandersjöö A, Maegele M, Bellander BM. Does Complement-Mediated Hemostatic Disturbance Occur in Traumatic Brain Injury? A Literature Review and Observational Study Protocol. Int J Mol Sci 2020; 21:E1596. [PMID: 32111078 PMCID: PMC7084711 DOI: 10.3390/ijms21051596] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 02/07/2020] [Accepted: 02/25/2020] [Indexed: 12/11/2022] Open
Abstract
Despite improvements in medical triage and tertiary care, traumatic brain injury (TBI) remains associated with significant morbidity and mortality. Almost two-thirds of patients with severe TBI develop some form of hemostatic disturbance, which contributes to poor outcome. In addition, the complement system, which is abundant in the healthy brain, undergoes significant intra- and extracranial amplification following TBI. Previously considered to be structurally similar but separate systems, evidence of an interaction between the complement and coagulation systems in non-TBI cohorts has accumulated, with the activation of one system amplifying the activation of the other, independent of their established pathways. However, it is not known whether this interaction exists in TBI. In this review we summarize the available literature on complement activation following TBI, and the crosstalk between the complement and coagulation systems. We demonstrate how the complement system interacts with the coagulation cascade by activating the intrinsic coagulation pathway and by bypassing the initial cascade and directly producing thrombin as well. This crosstalk also effects platelets, where evidence points to a relationship with the complement system on multiple levels, with complement anaphylatoxins being able to induce disproportionate platelet activation and adhesion. The complement system also stimulates thrombosis by inhibiting fibrinolysis and stimulating endothelial cells to release prothrombotic microparticles. These interactions see clinical relevance in several disorders where a deficiency in complement regulation seems to result in a prothrombotic clinical presentation. Finally, based on these observations, we present the outline of an observational cohort study that is currently under preparation and aimed at assessing how complement influences coagulation in patients with isolated TBI.
Collapse
Affiliation(s)
- Alexander Fletcher-Sandersjöö
- Department of Neurosurgery, Karolinska University Hospital, 171 76 Stockholm, Sweden;
- Department of Clinical Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Marc Maegele
- Department for Trauma and Orthopedic Surgery, Cologne-Merheim Medical Center (CMMC), University Witten/Herdecke, 58455 Cologne, Germany;
- Institute for Research in Operative Medicine, University Witten/Herdecke, 58455 Cologne, Germany
| | - Bo-Michael Bellander
- Department of Neurosurgery, Karolinska University Hospital, 171 76 Stockholm, Sweden;
- Department of Clinical Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden
| |
Collapse
|
117
|
van Gent JAN, van Essen TA, Bos MHA, Cannegieter SC, van Dijck JTJM, Peul WC. Coagulopathy after hemorrhagic traumatic brain injury, an observational study of the incidence and prognosis. Acta Neurochir (Wien) 2020; 162:329-336. [PMID: 31741112 PMCID: PMC6982633 DOI: 10.1007/s00701-019-04111-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 10/14/2019] [Indexed: 01/28/2023]
Abstract
Background Traumatic brain injury is associated with high rates of mortality and morbidity. Trauma patients with a coagulopathy have a 10-fold increased mortality risk compared to patients without a coagulopathy. The aim of this study was to identify the incidence of coagulopathy and relate early coagulopathy to clinical outcome in patients with traumatic intracranial hemorrhages. Methods Between September 2015 and December 2016, 108 consecutive cranial trauma patients with traumatic intracranial hemorrhages were included in this study. To assess the relationship between patients with a coagulopathy and outcome, a chi-squared test was performed. Results A total of 29 out of the 108 patients (27%) with a traumatic intracranial hemorrhage developed a coagulopathy within 72 h after admission. Overall, a total of 22 patients (20%) died after admission of which ten were coagulopathic at emergency department presentation. Early coagulopathy in patients with traumatic brain injury is associated with progression of hemorrhagic injury (odds ratio 2.4 (95% confidence interval 0.8–8.0)), surgical intervention (odds ratio 2.8 (95% confidence interval 0.87–9.35)), and increased in-hospital mortality (odds ratio 23.06 (95% confidence interval 5.5–95.9)). Conclusion Patients who sustained a traumatic intracranial hemorrhage remained at risk for developing a coagulopathy until 72 h after trauma. Patients who developed a coagulopathy had a worse clinical outcome than patients who did not develop a coagulopathy.
Collapse
Affiliation(s)
- Jort A. N. van Gent
- Department of Neurosurgery, Leiden University Medical Center, University Neurosurgical Center Holland (UNCH), Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- HAGA Teaching Hospital, The Hague, The Netherlands
- Haaglanden Medical Center, The Hague, The Netherlands
| | - Thomas A. van Essen
- Department of Neurosurgery, Leiden University Medical Center, University Neurosurgical Center Holland (UNCH), Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- HAGA Teaching Hospital, The Hague, The Netherlands
- Haaglanden Medical Center, The Hague, The Netherlands
| | - Mettine H. A. Bos
- Division of Thrombosis and Hemostasis, Leiden University Medical Center, Leiden, The Netherlands
| | - Suzanne C. Cannegieter
- Division of Thrombosis and Hemostasis, Leiden University Medical Center, Leiden, The Netherlands
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jeroen T. J. M. van Dijck
- Department of Neurosurgery, Leiden University Medical Center, University Neurosurgical Center Holland (UNCH), Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- HAGA Teaching Hospital, The Hague, The Netherlands
- Haaglanden Medical Center, The Hague, The Netherlands
| | - Wilco C. Peul
- Department of Neurosurgery, Leiden University Medical Center, University Neurosurgical Center Holland (UNCH), Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- HAGA Teaching Hospital, The Hague, The Netherlands
- Haaglanden Medical Center, The Hague, The Netherlands
| |
Collapse
|
118
|
Hanna K, Hamidi M, Anderson KT, Ditillo M, Zeeshan M, Tang A, Henry M, Kulvatunyou N, Joseph B. Pediatric resuscitation: Weight-based packed red blood cell volume is a reliable predictor of mortality. J Trauma Acute Care Surg 2020; 87:356-363. [PMID: 31349349 DOI: 10.1097/ta.0000000000002305] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND The definition of massive transfusion (MT) in civilian pediatric trauma patients is not established. In combat-injured pediatric patients, the definition of MT is based on the volume of total blood products transfused. The aim of this study is to define MT in civilian pediatric trauma patients based on a packed red blood cell (PRBC) volume threshold and compare its predictive power to a total blood products volume threshold. METHODS An analysis of the pediatric American College of Surgeons Trauma Quality Improvement Program database was performed (2014-2016) including pediatric trauma patients (4-18 years) who received blood products within 24 hours. Receiver operator characteristic curves for predicting mortality determined the optimal PRBC MT threshold. Area under receiver operating characteristic curve (AUROC) curve analysis was performed to compare the predictive power of a PRBC threshold to a total blood product threshold. RESULTS A total of 1,495 patients were included. Sensitivity and specificity for 24-hour and in-hospital mortality were optimal at a PRBC threshold of 20 mL/kg. As compared with total blood products threshold, 20 mL/kg PRBCs volume achieved higher discriminatory power for predicting 24-hour (AUROC, 0.803 vs. 0.672; p < 0.001) and in-hospital mortality (AUROC, 0.815 vs. 0.686, p < 0.001). Patients who received an MT had higher Injury Severity Score (p < 0.001) and were more likely to receive mechanical ventilation (p < 0.001) and intensive care unit admission (p < 0.001). Overall 24-hour mortality (23.1% vs. 7.6%, p < 0.001) and in-hospital mortality (44.9% vs. 15.8%, p < 0.001) were higher in the MT group. On regression analysis, MT significantly predicted in-hospital mortality (odds ratio, 3.8 [2.9-4.9, 95% CI]) and 24-hour mortality (odds ratio, 3.3 [2.4-4.7, 95% CI]). CONCLUSION The use of a PRBCs MT definition in civilian pediatric patients is a better predictor of mortality compared with total blood products threshold. These results provide a framework for MT protocol development. LEVEL OF EVIDENCE Prognostic study, level III.
Collapse
Affiliation(s)
- Kamil Hanna
- From the Division of Trauma, Critical Care, Emergency Surgery, and Burns, Department of Surgery (K.N., M.H., K.T.A., M.Z., A.T., M.H., N.K., B.J.), College of Medicine, University of Arizona, Tucson, Arizona; and Department of Trauma Surgery (M.D.), Allegheny General Hospital, Pittsburgh, Pennsylvania
| | | | | | | | | | | | | | | | | |
Collapse
|
119
|
Jost D, Lemoine S, Lemoine F, Lanoe V, Maurin O, Derkenne C, Franchin Frattini M, Delacote M, Seguineau E, Godefroy A, Hervault N, Delhaye L, Pouliquen N, Louis-Delauriere E, Trichereau J, Roquet F, Salomé M, Verret C, Bihannic R, Jouffroy R, Frattini B, Hong Tuan Ha V, Dang-Minh P, Travers S, Bignand M, Martinaud C, Garrabe E, Ausset S, Prunet B, Sailliol A, Tourtier JP. French lyophilized plasma versus normal saline for post-traumatic coagulopathy prevention and correction: PREHO-PLYO protocol for a multicenter randomized controlled clinical trial. Trials 2020; 21:106. [PMID: 31969168 PMCID: PMC6977230 DOI: 10.1186/s13063-020-4049-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 01/06/2020] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Post-trauma bleeding induces an acute deficiency in clotting factors, which promotes bleeding and hemorrhagic shock. However, early plasma administration may reduce the severity of trauma-induced coagulopathy (TIC). Unlike fresh frozen plasma, which requires specific hospital logistics, French lyophilized plasma (FLYP) is storable at room temperature and compatible with all blood types, supporting its use in prehospital emergency care. We aim to test the hypothesis that by attenuating TIC, FLYP administered by prehospital emergency physicians would benefit the severely injured civilian patient at risk for hemorrhagic shock. METHODS/DESIGN This multicenter randomized clinical trial will include adults severely injured and at risk for hemorrhagic shock, with a systolic blood pressure < 70 mmHg or a Shock Index > 1.1. Two parallel groups of 70 patients will receive either FLYP or normal saline in addition to usual treatment. The primary endpoint is the International Normalized Ratio (INR) at hospital admission. Secondary endpoints are transfusion requirement, length of stay in the intensive care unit, survival rate at day 30, usability and safety related to FLYP use, and other biological coagulation parameters. CONCLUSION With this trial, we aim to confirm the efficacy of FLYP in TIC and its safety in civilian prehospital care. The study results will contribute to optimizing guidelines for treating hemorrhagic shock in civilian settings. TRIAL REGISTRATION ClinicalTrials.gov, NCT02736812. Registered on 13 April 2016. The trial protocol has been approved by the French ethics committee (CPP 3342) and the French Agency for the Safety of Medicines and Health Products (IDRCB 2015-A00866-43).
Collapse
Affiliation(s)
- Daniel Jost
- Paris Fire Brigade Medical Emergency Department, 1 place Jules Renard, 75017, Paris, France.
| | - Sabine Lemoine
- Paris Fire Brigade Medical Emergency Department, 1 place Jules Renard, 75017, Paris, France
| | - Frederic Lemoine
- Paris Fire Brigade Medical Emergency Department, 1 place Jules Renard, 75017, Paris, France
| | - Vincent Lanoe
- Paris Fire Brigade Medical Emergency Department, 1 place Jules Renard, 75017, Paris, France
| | - Olga Maurin
- Paris Fire Brigade Medical Emergency Department, 1 place Jules Renard, 75017, Paris, France
| | - Clément Derkenne
- Paris Fire Brigade Medical Emergency Department, 1 place Jules Renard, 75017, Paris, France
| | | | - Maëlle Delacote
- Paris Fire Brigade Medical Emergency Department, 1 place Jules Renard, 75017, Paris, France
| | - Edouard Seguineau
- Paris Fire Brigade Medical Emergency Department, 1 place Jules Renard, 75017, Paris, France
| | - Anne Godefroy
- Paris Fire Brigade Medical Emergency Department, 1 place Jules Renard, 75017, Paris, France
| | - Nicolas Hervault
- Paris Fire Brigade Medical Emergency Department, 1 place Jules Renard, 75017, Paris, France
| | - Ludovic Delhaye
- Paris Fire Brigade Medical Emergency Department, 1 place Jules Renard, 75017, Paris, France
| | - Nicolas Pouliquen
- Paris Fire Brigade Medical Emergency Department, 1 place Jules Renard, 75017, Paris, France
| | - Emilie Louis-Delauriere
- Department of Education, Research and Innovation, Service de Santé des Armées, 1 Place Alphonse Laveran, 75230, Paris, France
| | - Julie Trichereau
- Paris Fire Brigade Medical Emergency Department, 1 place Jules Renard, 75017, Paris, France
| | - Florian Roquet
- Paris Fire Brigade Medical Emergency Department, 1 place Jules Renard, 75017, Paris, France
| | - Marina Salomé
- Paris Fire Brigade Medical Emergency Department, 1 place Jules Renard, 75017, Paris, France
| | - Catherine Verret
- Department of Education, Research and Innovation, Service de Santé des Armées, 1 Place Alphonse Laveran, 75230, Paris, France
| | - René Bihannic
- Paris Fire Brigade Medical Emergency Department, 1 place Jules Renard, 75017, Paris, France
| | - Romain Jouffroy
- Paris Fire Brigade Medical Emergency Department, 1 place Jules Renard, 75017, Paris, France
| | - Benoit Frattini
- Paris Fire Brigade Medical Emergency Department, 1 place Jules Renard, 75017, Paris, France
| | - Vivien Hong Tuan Ha
- Paris Fire Brigade Medical Emergency Department, 1 place Jules Renard, 75017, Paris, France
| | - Pascal Dang-Minh
- Paris Fire Brigade Medical Emergency Department, 1 place Jules Renard, 75017, Paris, France
| | - Stéphane Travers
- Paris Fire Brigade Medical Emergency Department, 1 place Jules Renard, 75017, Paris, France
- French Military Health Service, Val de Grâce military hospital, 1, Place Alphonse Laveran, 75230, Paris, France
| | - Michel Bignand
- Paris Fire Brigade Medical Emergency Department, 1 place Jules Renard, 75017, Paris, France
| | - Christophe Martinaud
- French Military Health Service, Val de Grâce military hospital, 1, Place Alphonse Laveran, 75230, Paris, France
- French army blood transfusion center, 1 Rue du Lieutenant Raoul Batany, 92140, Clamart, France
| | - Eliane Garrabe
- French Military Health Service, Val de Grâce military hospital, 1, Place Alphonse Laveran, 75230, Paris, France
- French army blood transfusion center, 1 Rue du Lieutenant Raoul Batany, 92140, Clamart, France
| | - Sylvain Ausset
- French Military Health Service, Val de Grâce military hospital, 1, Place Alphonse Laveran, 75230, Paris, France
- Department of Anesthesiology and Intensive Care, Percy military teaching hospital, 101 avenue Henri Barbusse, BP 406, 92141, Clamart, Cedex, France
| | - Bertrand Prunet
- Paris Fire Brigade Medical Emergency Department, 1 place Jules Renard, 75017, Paris, France
- French Military Health Service, Val de Grâce military hospital, 1, Place Alphonse Laveran, 75230, Paris, France
| | - Anne Sailliol
- French Military Health Service, Val de Grâce military hospital, 1, Place Alphonse Laveran, 75230, Paris, France
- French army blood transfusion center, 1 Rue du Lieutenant Raoul Batany, 92140, Clamart, France
- French Military Research Institute, 1 place Valérie Andre, BP 73, 91223, Brétigny sur Orge, France
| | - Jean Pierre Tourtier
- Paris Fire Brigade Medical Emergency Department, 1 place Jules Renard, 75017, Paris, France
- French Military Health Service, Val de Grâce military hospital, 1, Place Alphonse Laveran, 75230, Paris, France
- Department of Anaesthesiology and Intensive Care, Begin military teaching hospital, 94160, Saint-Mande, France
| |
Collapse
|
120
|
Dias JD, Lopez-Espina CG, Bliden K, Gurbel P, Hartmann J, Achneck HE. TEG®6s system measures the contributions of both platelet count and platelet function to clot formation at the site-of-care. Platelets 2019; 31:932-938. [PMID: 31878831 DOI: 10.1080/09537104.2019.1704713] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Knowledge of platelet count and function is key to ensuring appropriate hemostatic management. We hypothesized that the novel, portable TEG®6s coagulation assessment system could evaluate the contribution of both platelet count and function to clot formation. Whole-blood samples with variable platelet counts were prepared from healthy volunteers. Platelet function was adjusted using seven concentrations of abciximab and evaluated by light transmission aggregometry (LTA) with TRAP agonist. Maximum amplitude (MA), reaction time (R) and activated clotting time (ACT) were assessed in citrated kaolin (CK), CK with heparinase (CKH), citrated RapidTEG® (CRT), and citrated functional fibrinogen (CFF) assays. Positive correlations were observed between platelet count and CK.MA, CKH.MA, and CRT.MA (p < .0001), and CK.R, CKH.R, and CRT.ACT (p < .05). Platelet count could be accurately quantified in the range 28-91 k/μL, 28-86 k/μL and 28-74 k/μL for CK.MA, CKH.MA, and CRT.MA, respectively. CK.MA, CKH.MA, and CRT.MA showed significant negative relationships with abciximab concentration (p < .001). Platelet function inhibition was detected by all three assays at >68% measured by LTA and quantified in the range 68.4-82% (CK), 69.4-88% (CKH), and 69.7-76% (CRT). This demonstrates the TEG®6s analyzer can accurately evaluate platelet count and function at the site-of-care.
Collapse
Affiliation(s)
- Joao D Dias
- Clinical Development and Medical Affairs, Haemonetics Corporation , Signy, Switzerland
| | - Carlos G Lopez-Espina
- Clinical Development and Medical Affairs, Haemonetics Corporation , Rosemont, IL, USA
| | - Kevin Bliden
- Sinai Center for Thrombosis Research Sinai Hospital, 2401 W. Belvedere Ave, Baltimore MD 21215
| | - Paul Gurbel
- Sinai Center for Thrombosis Research Sinai Hospital, 2401 W. Belvedere Ave, Baltimore MD 21215
| | - Jan Hartmann
- Clinical Development and Medical Affairs, Haemonetics Corporation , Braintree, MA, USA
| | - Hardean E Achneck
- Clinical Development and Medical Affairs, Haemonetics Corporation , Braintree, MA, USA
| |
Collapse
|
121
|
Lopez E, Srivastava AK, Burchfield J, Wang YW, Cardenas JC, Togarrati PP, Miyazawa B, Gonzalez E, Holcomb JB, Pati S, Wade CE. Platelet-derived- Extracellular Vesicles Promote Hemostasis and Prevent the Development of Hemorrhagic Shock. Sci Rep 2019; 9:17676. [PMID: 31776369 PMCID: PMC6881357 DOI: 10.1038/s41598-019-53724-y] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 11/04/2019] [Indexed: 01/01/2023] Open
Abstract
Every year more than 500,000 deaths are attributed to trauma worldwide and severe hemorrhage is present in most of them. Transfused platelets have been shown to improve survival in trauma patients, although its mechanism is only partially known. Platelet derived-extracellular vesicles (PEVs) are small vesicles released from platelets upon activation and/or mechanical stimulation and many of the benefits attributed to platelets could be mediated through PEVs. Based on the available literature, we hypothesized that transfusion of human PEVs would promote hemostasis, reduce blood loss and attenuate the progression to hemorrhagic shock following severe trauma. In this study, platelet units from four different donors were centrifuged to separate platelets and PEVs. The pellets were washed to obtain plasma-free platelets to use in the rodent model. The supernatant was subjected to tangential flow filtration for isolation and purification of PEVs. PEVs were assessed by total count and particle size distribution by Nanoparticle Tracking Analysis (NTA) and characterized for cells of origin and expression of EV specific-surface and cytosolic markers by flow cytometry. The coagulation profile from PEVs was assessed by calibrated automated thrombography (CAT) and thromboelastography (TEG). A rat model of uncontrolled hemorrhage was used to compare the therapeutic effects of 8.7 × 108 fresh platelets (FPLT group, n = 8), 7.8 × 109 PEVs (PEV group, n = 8) or Vehicle (Control, n = 16) following severe trauma. The obtained pool of PEVs from 4 donors had a mean size of 101 ± 47 nm and expressed the platelet-specific surface marker CD41 and the EV specific markers CD9, CD61, CD63, CD81 and HSP90. All PEV isolates demonstrated a dose-dependent increase in the rate and amount of thrombin generated and overall clot strength. In vivo experiments demonstrated a 24% reduction in abdominal blood loss following liver trauma in the PEVs group when compared with the control group (9.9 ± 0.4 vs. 7.5 ± 0.5 mL, p < 0.001>). The PEV group also exhibited improved outcomes in blood pressure, lactate level, base excess and plasma protein concentration compared to the Control group. Fresh platelets failed to improve these endpoints when compared to Controls. Altogether, these results indicate that human PEVs provide pro-hemostatic support following uncontrolled bleeding. As an additional therapeutic effect, PEVs improve the outcome following severe trauma by maintaining hemodynamic stability and attenuating the development of ischemia, base deficit, and cardiovascular shock.
Collapse
Affiliation(s)
- Ernesto Lopez
- Center for Translational Injury Research (CeTIR), Department of Surgery, University of Texas Health Science Center at Houston, Houston, McGovern Medical School, Houston, TX, USA.
| | - Amit K Srivastava
- Department of Pediatric Surgery, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - John Burchfield
- Center for Translational Injury Research (CeTIR), Department of Surgery, University of Texas Health Science Center at Houston, Houston, McGovern Medical School, Houston, TX, USA
| | - Yao-Wei Wang
- Center for Translational Injury Research (CeTIR), Department of Surgery, University of Texas Health Science Center at Houston, Houston, McGovern Medical School, Houston, TX, USA
| | - Jessica C Cardenas
- Center for Translational Injury Research (CeTIR), Department of Surgery, University of Texas Health Science Center at Houston, Houston, McGovern Medical School, Houston, TX, USA
| | | | - Byron Miyazawa
- Department of Laboratory Medicine, University of California, San Francisco, CA, USA
| | - Erika Gonzalez
- Center for Translational Injury Research (CeTIR), Department of Surgery, University of Texas Health Science Center at Houston, Houston, McGovern Medical School, Houston, TX, USA
| | - John B Holcomb
- Center for Translational Injury Research (CeTIR), Department of Surgery, University of Texas Health Science Center at Houston, Houston, McGovern Medical School, Houston, TX, USA
| | - Shibani Pati
- Department of Laboratory Medicine, University of California, San Francisco, CA, USA
| | - Charles E Wade
- Center for Translational Injury Research (CeTIR), Department of Surgery, University of Texas Health Science Center at Houston, Houston, McGovern Medical School, Houston, TX, USA
| |
Collapse
|
122
|
Platelet Contributions to Trauma-Induced Coagulopathy: Updates in Post-injury Platelet Biology, Platelet Transfusions, and Emerging Platelet-Based Hemostatic Agents. CURRENT TRAUMA REPORTS 2019. [DOI: 10.1007/s40719-019-00176-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
123
|
Cudjoe EK, Hassan ZH, Kang L, Reynolds PS, Fisher BJ, McCarter J, Sweeney C, Martin EJ, Middleton P, Ellenberg M, Fowler AA, Spiess BD, Brophy DF, Hawkridge AM, Natarajan R. Temporal map of the pig polytrauma plasma proteome with fluid resuscitation and intravenous vitamin C treatment. J Thromb Haemost 2019; 17:1827-1837. [PMID: 31322812 DOI: 10.1111/jth.14580] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 07/16/2019] [Indexed: 01/12/2023]
Abstract
BACKGROUND Fluid resuscitation plays a prominent role in stabilizing trauma patients with hemorrhagic shock yet there remains uncertainty with regard to optimal administration time, volume, and fluid composition (e.g., whole blood, component, colloids) leading to complications such as trauma-induced coagulopathies (TIC), acidosis, and poor oxygen transport. Synthetic fluids in combination with antioxidants (e.g., vitamin C) may resolve some of these problems. OBJECTIVES We applied quantitative mass spectrometry-based proteomics [liquid chromatography-mass spectrometry (LC-MS/MS)] to map the effects of fluid resuscitation and intravenous vitamin C (VitC) in a pig model of polytrauma (hemorrhagic shock, tissue injury, liver reperfusion, hypothermia, and comminuted bone fracture). The goal was to determine the effects of VitC on plasma protein expression, with respect to changes associated with coagulation and trauma-induced coagulopathy (TIC). METHODS Longitudinal blood samples were drawn from nine male Sinclair pigs at baseline, 2 h post trauma, and 0.25, 2, and 4 h post fluid resuscitation with 500 mL hydroxyethyl starch. Pigs were treated intravenously (N = 3/treatment group) with saline, 50 mg VitC/kg (Lo-VitC), or 200 mg VitC/kg (Hi-VitC) during fluid resuscitation. RESULTS A total of 436 plasma proteins were quantified of which 136 changed following trauma and resuscitation; 34 were associated with coagulation, complement cascade, and glycolysis. Unexpectedly, Lo-VitC and Hi-VitC treatments stabilized ADAMTS13 levels by ~4-fold (P = .056) relative to saline and enhanced ADAMTS13/von Willebrand factor (VWF) cleavage efficiency based on LC-MS/MS evidence for the semitryptic VWF cleavage product (VWF1275-1286 ). CONCLUSIONS This study provides the first comprehensive map of trauma-induced changes to the plasma proteome, especially with respect to proteins driving the development of TIC.
Collapse
Affiliation(s)
- Emmanuel K Cudjoe
- Department of Pharmacotherapy and Outcomes Science, Virginia Commonwealth University School of Pharmacy, Richmond, Virginia
| | - Zaneera H Hassan
- Department of Pharmaceutics, Virginia Commonwealth University School of Pharmacy, Richmond, Virginia
| | - Le Kang
- Department of Biostatistics, Virginia Commonwealth University Medical Center, Richmond, Virginia
| | - Penny S Reynolds
- Department of Anesthesiology, Virginia Commonwealth University Medical Center, Richmond, Virginia
| | - Bernard J Fisher
- Department of Internal Medicine, Virginia Commonwealth University Medical Center, Richmond, Virginia
| | - Jacquelyn McCarter
- Department of Internal Medicine, Virginia Commonwealth University Medical Center, Richmond, Virginia
| | - Christopher Sweeney
- Department of Internal Medicine, Virginia Commonwealth University Medical Center, Richmond, Virginia
| | - Erika J Martin
- Department of Pharmacotherapy and Outcomes Science, Virginia Commonwealth University School of Pharmacy, Richmond, Virginia
| | - Paul Middleton
- Department of Internal Medicine, Virginia Commonwealth University Medical Center, Richmond, Virginia
| | - Matthew Ellenberg
- Department of Internal Medicine, Virginia Commonwealth University Medical Center, Richmond, Virginia
| | - Alpha A Fowler
- Department of Internal Medicine, Virginia Commonwealth University Medical Center, Richmond, Virginia
| | - Bruce D Spiess
- Department of Anesthesiology, Virginia Commonwealth University Medical Center, Richmond, Virginia
| | - Donald F Brophy
- Department of Pharmacotherapy and Outcomes Science, Virginia Commonwealth University School of Pharmacy, Richmond, Virginia
| | - Adam M Hawkridge
- Department of Pharmacotherapy and Outcomes Science, Virginia Commonwealth University School of Pharmacy, Richmond, Virginia
- Department of Pharmaceutics, Virginia Commonwealth University School of Pharmacy, Richmond, Virginia
- Institute for Structural Biology, Drug Discovery, and Development, Virginia Commonwealth University School of Pharmacy, Richmond, Virginia
| | - Ramesh Natarajan
- Department of Internal Medicine, Virginia Commonwealth University Medical Center, Richmond, Virginia
| |
Collapse
|
124
|
Huish S, Green L, Curnow E, Wiltshire M, Cardigan R. Effect of storage of plasma in the presence of red blood cells and platelets: re-evaluating the shelf life of whole blood. Transfusion 2019; 59:3468-3477. [PMID: 31618457 DOI: 10.1111/trf.15549] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 08/13/2019] [Accepted: 08/15/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND There is renewed interest in administering whole blood (WB) for the resuscitation of patients with bleeding trauma. The shelf life of WB was established decades ago based on the viability of red blood cells. However, plasma quality during WB storage is not established. STUDY DESIGN AND METHODS White blood cell- and platelet-reduced WB (WB-PLT) was prepared using standard processes and compared to WB processed using a platelet-sparing WBC reduction (WB + PLT) filter. WB (± PLT) was held at 2 to 6°C for 35 days alongside control units of red blood cells (RBCs) in saline, adenine, glucose, and mannitol and liquid plasma. A series of assays explored the coagulation potential and RBC quality. RESULTS While fibrinogen and α2-antiplasmin remained unaffected by storage, other factors varied between components or over time at 2 to 6°C. At 14 days factor V, factor VII, α2 -antiplasmin and free protein S antigen remained on average greater than 0.50 IU/mL or 50%, as appropriate, in WB ± PLT. Factor VIII was on average 0.49 IU/mL in WB+PLT, and 0.56 IU/mL for WB-PLT. Free protein S activity decreased significantly in all arms but remained on average greater than 40% at Day 14. Contact activation was not demonstrated before Day 14. Thrombin generation in plasma remained relatively stable to Day 35 in all arms. CONCLUSIONS Clotting factor activity remained at or above a mean of 0.5 IU/mL, or 50%, at Day 14 for factor V, factor VII, factor VIII, free protein S, fibrinogen, and α2-antiplasmin in all arms. Further data on platelet function in WB+PLT is needed to inform its shelf life.
Collapse
Affiliation(s)
| | - Laura Green
- NHS Blood and Transplant, London, UK.,Barts Health NHS Trust, London, UK.,Blizard Institute, Queen Mary University of London, London, UK
| | | | | | - Rebecca Cardigan
- NHS Blood and Transplant, Cambridge, UK.,Department of Haematology, University of Cambridge, Cambridge, UK
| |
Collapse
|
125
|
Girish A, Hickman DA, Banerjee A, Luc N, Ma Y, Miyazawa K, Sekhon UDS, Sun M, Huang S, Sen Gupta A. Trauma-targeted delivery of tranexamic acid improves hemostasis and survival in rat liver hemorrhage model. J Thromb Haemost 2019; 17:1632-1644. [PMID: 31220416 PMCID: PMC10124760 DOI: 10.1111/jth.14552] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 06/13/2019] [Accepted: 06/17/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND Trauma-associated hemorrhage and coagulopathy remain leading causes of mortality. Such coagulopathy often leads to a hyperfibrinolytic phenotype where hemostatic clots become unstable because of upregulated tissue plasminogen activator (tPA) activity. Tranexamic acid (TXA), a synthetic inhibitor of tPA, has emerged as a promising drug to mitigate fibrinolysis. TXA is US Food and Drug Administration-approved for treating heavy menstrual and postpartum bleeding, and has shown promise in trauma treatment. However, emerging reports also implicate TXA for off-target systemic coagulopathy, thromboembolic complications, and neuropathy. OBJECTIVE We hypothesized that targeted delivery of TXA to traumatic injury site can enable its clot-stabilizing action site-selectively, to improve hemostasis and survival while avoiding off-target effects. To test this, we used liposomes as a model delivery vehicle, decorated their surface with a fibrinogen-mimetic peptide for anchorage to active platelets within trauma-associated clots, and encapsulated TXA within them. METHODS The TXA-loaded trauma-targeted nanovesicles (T-tNVs) were evaluated in vitro in rat blood, and then in vivo in a liver trauma model in rats. TXA-loaded control (untargeted) nanovesicles (TNVs), free TXA, or saline were studied as comparison groups. RESULTS Our studies show that in vitro, the T-tNVs could resist lysis in tPA-spiked rat blood. In vivo, T-tNVs maintained systemic safety, significantly reduced blood loss and improved survival in the rat liver hemorrhage model. Postmortem evaluation of excised tissue from euthanized rats confirmed systemic safety and trauma-targeted activity of the T-tNVs. CONCLUSION Overall, the studies establish the potential of targeted TXA delivery for safe injury site-selective enhancement and stabilization of hemostatic clots to improve survival in trauma.
Collapse
Affiliation(s)
- Aditya Girish
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - DaShawn A. Hickman
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio
| | - Ankush Banerjee
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Norman Luc
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Yifeng Ma
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Kenji Miyazawa
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Ujjal D. S. Sekhon
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Michael Sun
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Stephanie Huang
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Anirban Sen Gupta
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
126
|
Risk factors for death and amputation in acute leg compartment syndrome. EUROPEAN JOURNAL OF ORTHOPAEDIC SURGERY AND TRAUMATOLOGY 2019; 30:359-365. [PMID: 31560102 DOI: 10.1007/s00590-019-02563-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Accepted: 09/24/2019] [Indexed: 12/29/2022]
Abstract
INTRODUCTION The primary objective of this study is to determine whether time from injury to fasciotomy is associated with increased risk for death or limb amputation in patients with acute leg compartment syndrome. The secondary objective of this study is to identify other risk factors for death or limb amputation in patients with acute leg compartment syndrome. METHODS In an institutional review board approved retrospective study, we identified 546 patients with acute compartment syndrome of 558 legs treated with fasciotomies from January 2000 to June 2015 at two Level I trauma centers. Our primary outcome measures were death and limb amputation during inpatient hospital admission. Electronic medical records were analyzed for patient-related factors and treatment-related factors. Bivariate analyses were used to screen for variables associated with our primary outcome measures, and explanatory variables with a p value below 0.05 were included in our multivariable logistic regression analyses. RESULTS In-hospital death occurred in 6.6% and in-hospital limb amputation occurred in 9.5% of acute leg compartment syndrome patients. Neither death nor limb amputation was found to be associated with time from injury to fasciotomy. Multivariable logistic regression analyses showed that older age (p = 0.03), higher modified Charlson Comorbidity Index (p = 0.009), higher potassium (p = 0.02), lower hemoglobin (p = 0.002), and higher lactate (p < 0.001) were associated with death, and diabetes mellitus (p = 0.05), no compartment pressure measurement (p = 0.009), higher PTT (p = 0.03), and lower albumin (p = 0.01) were associated with limb amputation. CONCLUSIONS Time to fasciotomy is not found to be associated with death or limb amputation in acute leg compartment syndrome. Death and limb amputation are associated with patient-related factors and injury severity. LEVEL OF EVIDENCE Level III Prognostic.
Collapse
|
127
|
Serchan P, Shorten G, Maher M, Power SP. Pulmonary embolism occurring early after major trauma. BMJ Case Rep 2019; 12:12/9/e228783. [PMID: 31540919 DOI: 10.1136/bcr-2018-228783] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Pulmonary embolism (PE) secondary to trauma is the third most common cause of death in trauma patients who have survived 24 hours following injury. We describe a case of PE diagnosed within 3 hours of a major trauma in a previously well adolescent female. The early occurrence of PE in this case is at odds with what is generally reported (3-5 days) after major trauma. General consensus is that patients who suffer major trauma move from an initial hypocoaguable state, with increased risk of bleeding, to normocoagulable or hypercoaguable state, with a subsequent increased risk of venothromboembolism. However, Sumislawski et al recently demonstrated that a marginally greater proportion of trauma patients were in fact hypercoaguable rather than hypocoaguable on arrival to hospital and that trauma-induced coagulopathy tended to resolve within 24 hours; such data cause us to re-evaluate when to commence thromboprophylaxis for major trauma patients.
Collapse
Affiliation(s)
| | - George Shorten
- Anaesthesia and Intensive Care, Cork University Hospital, Cork, Ireland
| | - Michael Maher
- Department of Radiology, University College Cork, Cork, Ireland
| | | |
Collapse
|
128
|
Prothrombin, alone or in complex concentrates or plasma, reduces bleeding in a mouse model of blood exchange-induced coagulopathy. Sci Rep 2019; 9:13029. [PMID: 31506556 PMCID: PMC6736877 DOI: 10.1038/s41598-019-49552-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 08/27/2019] [Indexed: 11/29/2022] Open
Abstract
Prothrombin complex concentrates (PCC) are fractionated plasma protein drugs that reverse warfarin anticoagulation. PCC may control more general bleeding. We sought to identify the dominant procoagulant factor in PCC in vivo. We tested PCC or coagulation factor (F) treatment in CD1 mice made coagulopathic by exchange of whole blood for washed red cells. Anesthetized mice were transfused with murine fresh-frozen plasma (mFFP), PCC, mixtures of human vitamin K-dependent proteins (VKDP) (prothrombin, FVII, FIX, or FX), or purified single human VKDP, immediately prior to tail transection (TT), liver laceration (LL), or intravascular laser injury (ILI). Plasma donor mice were treated with vehicle or control antisense oligonucleotide (ASO-CON) or ASO specific for prothrombin (FII) (ASO-FII) to yield mFFP or ASO-CON mFFP or ASO-FII mFFP. Blood losses were determined spectrophotometrically (TT) or gravimetrically (LL). Thrombus formation was quantified by intravital microscopy of laser-injured arterioles. PCC or four factor- (4F-) VKDP or prothrombin significantly reduced bleeding from TT or LL. Omission of prothrombin from 4F-VKDP significantly reduced its ability to limit bleeding. Mice transfused with ASO-FII mFFP demonstrated inferior haemostasis versus those transfused with ASO-FII following TT, LL, or ILI. Prothrombin is the dominant procoagulant component of PCC and could limit bleeding in trauma.
Collapse
|
129
|
Sheppard FR, Schaub LJ, Cap AP, Macko AR, Moore HB, Moore EE, Glaser CJJ. Whole blood mitigates the acute coagulopathy of trauma and avoids the coagulopathy of crystalloid resuscitation. J Trauma Acute Care Surg 2019; 85:1055-1062. [PMID: 30124622 DOI: 10.1097/ta.0000000000002046] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION The contributions of type and timing of fluid resuscitation to coagulopathy in trauma remain controversial. As part of a multifunctional resuscitation fluid research effort, we sought to further characterize the coagulation responses to resuscitation, specifically as compared to whole blood. We hypothesized that early whole blood administration mitigates the acute coagulopathy of trauma by avoiding the coagulopathy of CR resuscitation. METHODS Anesthetized rhesus macaques underwent polytraumatic, hemorrhagic shock, then a crossover study design resuscitation (n = 6 each) with either whole blood first (WB-1st) followed by crystalloid (CR); or CR-1st followed by WB. Resuscitation strategies were the following: WB-1st received 50% shed blood in 30minutes, followed by twice the shed blood volume (SBV) of CR over 30minutes and one times the SBV CR over 60minutes, where CR-1st received twice the SBV of CR over 30minutes, followed by 50% of shed blood in 30minutes, and one times the SBV CR over 60minutes. Blood samples were collected at baseline, end-of-shock, end-of-first and end-of-second resuscitation stages, and end-of-resuscitation for assessment (thromboelastometry, platelet aggregation, and plasmatic coagulation factors). Statistical analyses were conducted using two-way analysis of variance ANOVA with Bonferroni correction and t-tests; significance was at p < 0.05. RESULTS Survival, blood loss, hemodynamics, and shock duration were equivalent between the groups. Compared to baseline, parameters measured at first and second resuscitation stage time points directly following CR infusion revealed abnormalities in thromboelastometry (clot formation time, α angle, and maximum clot firmness), platelet aggregation response (to collagen, arachidonic acid, and adenosine diphosphate), and plasmatic coagulation (prothrombin time, anti-thrombin 3, and fibrinogen), while whole blood infusion resulted in stabilization or correction of these parameters following its administration. CONCLUSIONS These data suggest that in the setting of trauma and hemorrhagic shock, the coagulation alterations begin before intervention/resuscitation; however, these are significantly aggravated by CR resuscitation and could perhaps be best termed acute coagulopathy of resuscitation. STUDY TYPE Translational animal model.
Collapse
Affiliation(s)
- Forest R Sheppard
- From the Naval Medical Research Unit San Antonio, JBSA-Ft Sam Houston, Texas (F.R.S., L.J.S., A.R.M., J.J.G.); Maine Medical Center, Portland, Maine (F.R.S.); US Army Institute of Surgical Research, JBSA-Ft Sam Houston, Texas (A.P.C.); Department of Surgery, Denver Health Medical Center, Denver, Colorado (H.B.M., E.E.M); and University of Colorado Denver, Aurora, Colorado (H.B.M., E.E.M.)
| | | | | | | | | | | | | |
Collapse
|
130
|
Tsiklidis EJ, Sinno T, Diamond SL. Coagulopathy implications using a multiscale model of traumatic bleeding matching macro- and microcirculation. Am J Physiol Heart Circ Physiol 2019; 317:H73-H86. [PMID: 30978134 DOI: 10.1152/ajpheart.00774.2018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Quantifying the relationship between vascular injury and the dynamic bleeding rate requires a multiscale model that accounts for changing and coupled hemodynamics between the global and microvascular levels. A lumped, global hemodynamic model of the human cardiovascular system with baroreflex control was coupled to a local 24-level bifurcating vascular network that spanned diameters from the muscular artery scale (0.1-1.3 mm) to capillaries (5-10 μm) via conservation of momentum and conservation of mass boundary conditions. For defined injuries of severing all vessels at each nth-level, the changing pressures and flowrates were calculated using prescribed shear-dependent hemostatic clot growth rates (normal or coagulopathic). Key results were as follows: 1) the upstream vascular network rapidly depressurizes to reduce blood loss; 2) wall shear rates at the hemorrhaging wound exit are sufficiently high (~10,000 s-1) to drive von Willebrand factor unfolding; 3) full coagulopathy results in >2-liter blood loss in 2 h for severing all vessels of 0.13- to 0.005-mm diameter within the bifurcating network, whereas full hemostasis limits blood loss to <100 ml within 2 min; and 4) hemodilution from transcapillary refill increases blood loss and could be implicated in trauma-induced coagulopathy. A sensitivity analysis on length-to-diameter ratio and branching exponent demonstrated that bleeding was strongly dependent on these tissue-dependent network parameters. This is the first bleeding model that prescribes the geometry of the injury to calculate the rate of pressure-driven blood loss and local wall shear rate in the presence or absence of coagulopathic blood. NEW & NOTEWORTHY We developed a multiscale model that couples a lumped, global hemodynamic model of a patient to resolved, single-vessel wounds ranging from the small artery to capillary scale. The model is able to quantify wall shear rates, seal rates, and blood loss rates in the presence and absence of baroreflex control and hemodilution.
Collapse
Affiliation(s)
- Evan J Tsiklidis
- Department of Chemical and Biomolecular Engineering, Institute for Medicine and Engineering, University of Pennsylvania , Philadelphia, Pennsylvania
| | - Talid Sinno
- Department of Chemical and Biomolecular Engineering, Institute for Medicine and Engineering, University of Pennsylvania , Philadelphia, Pennsylvania
| | - Scott L Diamond
- Department of Chemical and Biomolecular Engineering, Institute for Medicine and Engineering, University of Pennsylvania , Philadelphia, Pennsylvania
| |
Collapse
|
131
|
Spahn DR, Bouillon B, Cerny V, Duranteau J, Filipescu D, Hunt BJ, Komadina R, Maegele M, Nardi G, Riddez L, Samama CM, Vincent JL, Rossaint R. The European guideline on management of major bleeding and coagulopathy following trauma: fifth edition. Crit Care 2019; 23:98. [PMID: 30917843 PMCID: PMC6436241 DOI: 10.1186/s13054-019-2347-3] [Citation(s) in RCA: 743] [Impact Index Per Article: 123.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 02/06/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Severe traumatic injury continues to present challenges to healthcare systems around the world, and post-traumatic bleeding remains a leading cause of potentially preventable death among injured patients. Now in its fifth edition, this document aims to provide guidance on the management of major bleeding and coagulopathy following traumatic injury and encourages adaptation of the guiding principles described here to individual institutional circumstances and resources. METHODS The pan-European, multidisciplinary Task Force for Advanced Bleeding Care in Trauma was founded in 2004, and the current author group included representatives of six relevant European professional societies. The group applied a structured, evidence-based consensus approach to address scientific queries that served as the basis for each recommendation and supporting rationale. Expert opinion and current clinical practice were also considered, particularly in areas in which randomised clinical trials have not or cannot be performed. Existing recommendations were re-examined and revised based on scientific evidence that has emerged since the previous edition and observed shifts in clinical practice. New recommendations were formulated to reflect current clinical concerns and areas in which new research data have been generated. RESULTS Advances in our understanding of the pathophysiology of post-traumatic coagulopathy have supported improved management strategies, including evidence that early, individualised goal-directed treatment improves the outcome of severely injured patients. The overall organisation of the current guideline has been designed to reflect the clinical decision-making process along the patient pathway in an approximate temporal sequence. Recommendations are grouped behind the rationale for key decision points, which are patient- or problem-oriented rather than related to specific treatment modalities. While these recommendations provide guidance for the diagnosis and treatment of major bleeding and coagulopathy, emerging evidence supports the author group's belief that the greatest outcome improvement can be achieved through education and the establishment of and adherence to local clinical management algorithms. CONCLUSIONS A multidisciplinary approach and adherence to evidence-based guidance are key to improving patient outcomes. If incorporated into local practice, these clinical practice guidelines have the potential to ensure a uniform standard of care across Europe and beyond and better outcomes for the severely bleeding trauma patient.
Collapse
Affiliation(s)
- Donat R. Spahn
- Institute of Anaesthesiology, University of Zurich and University Hospital Zurich, Raemistrasse 100, CH-8091 Zurich, Switzerland
| | - Bertil Bouillon
- Department of Trauma and Orthopaedic Surgery, Cologne-Merheim Medical Centre (CMMC), University of Witten/Herdecke, Ostmerheimer Strasse 200, D-51109 Cologne, Germany
| | - Vladimir Cerny
- Department of Anaesthesiology, Perioperative Medicine and Intensive Care, J.E. Purkinje University, Masaryk Hospital, Usti nad Labem, Socialni pece 3316/12A, CZ-40113 Usti nad Labem, Czech Republic
- Centre for Research and Development, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic, Sokolska 581, CZ-50005 Hradec Kralove, Czech Republic
- Department of Anaesthesiology and Intensive Care Medicine, Faculty of Medicine in Hradec Kralove, Charles University, Simkova 870, CZ-50003 Hradec Kralove, Czech Republic
- Department of Anaesthesia, Pain Management and Perioperative Medicine, QE II Health Sciences Centre, Dalhousie University, Halifax, 10 West Victoria, 1276 South Park St, Halifax, NS B3H 2Y9 Canada
| | - Jacques Duranteau
- Department of Anaesthesia and Intensive Care, Hôpitaux Universitaires Paris Sud, University of Paris XI, Faculté de Médecine Paris-Sud, 78 rue du Général Leclerc, F-94275 Le Kremlin-Bicêtre Cedex, France
| | - Daniela Filipescu
- Department of Cardiac Anaesthesia and Intensive Care, C. C. Iliescu Emergency Institute of Cardiovascular Diseases, Sos Fundeni 256-258, RO-022328 Bucharest, Romania
| | - Beverley J. Hunt
- King’s College and Departments of Haematology and Pathology, Guy’s and St Thomas’ NHS Foundation Trust, Westminster Bridge Road, London, SE1 7EH UK
| | - Radko Komadina
- Department of Traumatology, General and Teaching Hospital Celje, Medical Faculty Ljubljana University, SI-3000 Celje, Slovenia
| | - Marc Maegele
- Department of Trauma and Orthopaedic Surgery, Cologne-Merheim Medical Centre (CMMC), Institute for Research in Operative Medicine (IFOM), University of Witten/Herdecke, Ostmerheimer Strasse 200, D-51109 Cologne, Germany
| | - Giuseppe Nardi
- Department of Anaesthesia and ICU, AUSL della Romagna, Infermi Hospital Rimini, Viale Settembrini, 2, I-47924 Rimini, Italy
| | - Louis Riddez
- Department of Surgery and Trauma, Karolinska University Hospital, S-171 76 Solna, Sweden
| | - Charles-Marc Samama
- Hotel-Dieu University Hospital, 1, place du Parvis de Notre-Dame, F-75181 Paris Cedex 04, France
| | - Jean-Louis Vincent
- Department of Intensive Care, Erasme University Hospital, Université Libre de Bruxelles, Route de Lennik 808, B-1070 Brussels, Belgium
| | - Rolf Rossaint
- Department of Anaesthesiology, University Hospital Aachen, RWTH Aachen University, Pauwelsstrasse 30, D-52074 Aachen, Germany
| |
Collapse
|
132
|
Kim OV, Nevzorova TA, Mordakhanova ER, Ponomareva AA, Andrianova IA, Le Minh G, Daminova AG, Peshkova AD, Alber MS, Vagin O, Litvinov RI, Weisel JW. Fatal dysfunction and disintegration of thrombin-stimulated platelets. Haematologica 2019; 104:1866-1878. [PMID: 30792211 PMCID: PMC6717590 DOI: 10.3324/haematol.2018.202309] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 02/14/2019] [Indexed: 12/12/2022] Open
Abstract
Platelets play a key role in the formation of hemostatic clots and obstructive thrombi as well as in other biological processes. In response to physiological stimulants, including thrombin, platelets change shape, express adhesive molecules, aggregate, and secrete bioactive substances, but their subsequent fate is largely unknown. Here we examined late-stage structural, metabolic, and functional consequences of thrombin-induced platelet activation. Using a combination of confocal microscopy, scanning and transmission electron microscopy, flow cytometry, biochemical and biomechanical measurements, we showed that thrombin-induced activation is followed by time-dependent platelet dysfunction and disintegration. After ~30 minutes of incubation with thrombin, unlike with collagen or ADP, human platelets disintegrated into cellular fragments containing organelles, such as mitochondria, glycogen granules, and vacuoles. This platelet fragmentation was preceded by Ca2+ influx, integrin αIIbβ3 activation and phosphatidylserine exposure (activation phase), followed by mitochondrial depolarization, generation of reactive oxygen species, metabolic ATP depletion and impairment of platelet contractility along with dramatic cytoskeletal rearrangements, concomitant with platelet disintegration (death phase). Coincidentally with the platelet fragmentation, thrombin caused calpain activation but not activation of caspases 3 and 7. Our findings indicate that the late functional and structural damage of thrombin-activated platelets comprise a calpain-dependent platelet death pathway that shares some similarities with the programmed death of nucleated cells, but is unique to platelets, therefore representing a special form of cellular destruction. Fragmentation of activated platelets suggests that there is an underappreciated pathway of enhanced elimination of platelets from the circulation in (pro)thrombotic conditions once these cells have performed their functions.
Collapse
Affiliation(s)
- Oleg V Kim
- University of Pennsylvania Perelman School of Medicine, Department of Cell and Developmental Biology, Philadelphia, PA, USA.,University of California Riverside, Department of Mathematics, Riverside, CA, USA
| | - Tatiana A Nevzorova
- Kazan Federal University, Institute of Fundamental Medicine and Biology, Kazan, Russian Federation
| | - Elmira R Mordakhanova
- Kazan Federal University, Institute of Fundamental Medicine and Biology, Kazan, Russian Federation
| | - Anastasia A Ponomareva
- Kazan Federal University, Institute of Fundamental Medicine and Biology, Kazan, Russian Federation.,Kazan Institute of Biochemistry and Biophysics, FRC Kazan Scientific Center of RAS, Kazan, Russian Federation
| | - Izabella A Andrianova
- Kazan Federal University, Institute of Fundamental Medicine and Biology, Kazan, Russian Federation
| | - Giang Le Minh
- Kazan Federal University, Institute of Fundamental Medicine and Biology, Kazan, Russian Federation
| | - Amina G Daminova
- Kazan Federal University, Institute of Fundamental Medicine and Biology, Kazan, Russian Federation.,Kazan Institute of Biochemistry and Biophysics, FRC Kazan Scientific Center of RAS, Kazan, Russian Federation
| | - Alina D Peshkova
- Kazan Federal University, Institute of Fundamental Medicine and Biology, Kazan, Russian Federation
| | - Mark S Alber
- University of California Riverside, Department of Mathematics, Riverside, CA, USA
| | - Olga Vagin
- Geffen School of Medicine at UCLA, Department of Physiology, Los Angeles, CA, USA.,VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Rustem I Litvinov
- University of Pennsylvania Perelman School of Medicine, Department of Cell and Developmental Biology, Philadelphia, PA, USA.,Kazan Federal University, Institute of Fundamental Medicine and Biology, Kazan, Russian Federation
| | - John W Weisel
- University of Pennsylvania Perelman School of Medicine, Department of Cell and Developmental Biology, Philadelphia, PA, USA
| |
Collapse
|
133
|
Fluid Management and Transfusion. Int Anesthesiol Clin 2019; 55:78-95. [PMID: 28598882 DOI: 10.1097/aia.0000000000000154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
134
|
Abstract
Microparticles are submicron vesicles shed from aging erythrocytes as a characteristic feature of the red blood cell (RBC) storage lesion. Exposure of pulmonary endothelial cells to RBC-derived microparticles promotes an inflammatory response, but the mechanisms underlying microparticle-induced endothelial cell activation are poorly understood. In the present study, cultured murine lung endothelial cells (MLECs) were treated with microparticles isolated from aged murine packed RBCs or vehicle. Microparticle-treated cells demonstrated increased expression of the adhesion molecules ICAM and E-selectin, as well as the cytokine, IL-6. To identify mechanisms that mediate these effects of microparticles on MLECs, cells were treated with microparticles covalently bound to carboxyfluorescein succinimidyl ester (CFSE) and cellular uptake of microparticles was quantified via flow cytometry. Compared with controls, there was a greater proportion of CFSE-positive MLECs from 15 min up to 24 h, suggesting endocytosis of the microparticles by endothelial cells. Colocalization of microparticles with lysosomes was observed via immunofluorescence, indicating endocytosis and endolysosomal trafficking. This process was inhibited by endocytosis inhibitors. SiRNA knockdown of Rab5 signaling protein in endothelial cells resulted in impaired microparticle uptake as compared with nonsense siRNA-treated cells, as well as an attenuation of the inflammatory response to microparticle treatment. Taken together, these data suggest that endocytosis of RBC-derived microparticles by lung endothelial cells results in endothelial cell activation. This response seems to be mediated, in part, by the Rab5 signaling protein.
Collapse
|
135
|
Effects of in-house cryoprecipitate on transfusion usage and mortality in patients with multiple trauma with severe traumatic brain injury: a retrospective cohort study. BLOOD TRANSFUSION = TRASFUSIONE DEL SANGUE 2019; 18:6-12. [PMID: 30747700 DOI: 10.2450/2019.0198-18] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 12/14/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND Hypofibrinogenaemia is a common complication of multiple trauma with severe traumatic brain injury (Abbreviated Injury Scale score of the head ≥4; body ≥3). In Japan, neither fibrinogen concentrate nor cryoprecipitate is permitted to treat acquired hypofibrinogenaemia with the purpose of rapidly restoring a haemostatic level of fibrinogen. The aim of this study was to investigate transfusion usage and mortality in patients with multiple trauma and severe traumatic brain injury who were given a cryoprecipitate prepared in-house, comparing those administered the product early or later. MATERIAL AND METHODS We prepared and produced cryoprecipitate from fresh-frozen plasma beginning in March 2013. We performed a retrospective cohort study of patients admitted to our single tertiary medical centre with severe multiple trauma with traumatic brain injury from March 2013 to June 2018, sorting them into those given the cryoprecipitate infusion within 90 minutes of admission (Early group) and those given it more than 90 minutes after admission (Late group). Clinical outcomes were compared between the two groups using chi-square or Fisher's exact tests and the Wilcoxon test as appropriate. RESULTS There were 26 and 16 patients in the Early and Late groups, respectively. The 24-hour mortality tended to be lower in the Early group than in the Late group (8 vs 13%, respectively). The patients were more severely anaemic and thrombocytopenic after haemostatic therapy in the Late group than in the Early group. Transfusion usage in the Early group was lower than that in the Late group (red blood cells: 7±1 units vs 17±3 units, p<0.05; fresh-frozen plasma: 9±1 units vs 16±3 units, p<0.05; platelet concentrate: 3±1 units vs 15±4 units, p<0.05, respectively). DISCUSSION Early administration of an in-house cryoprecipitate may reduce transfusion usage in patients with multiple trauma with severe traumatic brain injury.
Collapse
|
136
|
Abstract
Uncontrolled bleeding is the leading preventable cause of death in patients with multiple injuries. Currently, trauma-induced coagulopathy is seen as an independent disease entity influencing survival. Severely bleeding trauma patients are often treated with classical blood products in predefined ratios (damage control resuscitation). Viscoelasticity-based and target-oriented approaches could possibly be given priority. Viscoelasticity-based diagnostics and therapy enable the qualitative investigation of whole blood and provide therapeutically usable information on initiation, dynamics and sustainability of thrombus formation. Due to the ease of handling and timely results this lends itself as a point-of-care procedure. This article presents the clinical issues with using viscoelastic procedures and current expert recommendations taking the literature into consideration.
Collapse
|
137
|
Longstaff C, Locke M. Increased urokinase and consumption of α 2 -antiplasmin as an explanation for the loss of benefit of tranexamic acid after treatment delay. J Thromb Haemost 2019; 17:195-205. [PMID: 30451372 PMCID: PMC6334274 DOI: 10.1111/jth.14338] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Indexed: 12/26/2022]
Abstract
Essentials Delayed treatment with tranexamic acid results in loss of efficacy and poor outcomes. Increasing urokinase activity may account for adverse effects of late tranexamic acid treatment. Urokinase + tranexamic acid produces plasmin in plasma or blood and disrupts clotting. α2 -Antiplasmin consumption with ongoing fibrinolysis increases plasmin-induced coagulopathy. SUMMARY: Background Tranexamic acid (TXA) is an effective antifibrinolytic agent with a proven safety record. However, large clinical trials show TXA becomes ineffective or harmful if treatment is delayed beyond 3 h. The mechanism is unknown but urokinase plasminogen activator (uPA) has been implicated. Methods Inhibitory mechanisms of TXA were explored in a variety of clot lysis systems using plasma and whole blood. Lysis by tissue plasminogen activator (tPA), uPA and plasmin were investigated. Coagulopathy was investigated using ROTEM and activated partial thromboplastin time (APTT). Results IC50 values for antifibrinolytic activity of TXA varied from < 10 to > 1000 μmol L-1 depending on the system, but good fibrin protection was observed in the presence of tPA, uPA and plasmin. However, in plasma or blood, active plasmin was generated by TXA + uPA (but not tPA) and coagulopathy developed leading to no or poor clot formation. The extent of coagulopathy was sensitive to available α2 -antiplasmin. No clot formed with plasma containing 40% normal α2 -antiplasmin after short incubation with TXA + uPA. Adding purified α2 -antiplasmin progressively restored clotting. Plasmin could be inhibited by aprotinin, IC50 = 530 nmol L-1 , in plasma. Conclusions Tranexamic acid protects fibrin but stimulates uPA activity and slows inhibition of plasmin by α2 -antiplasmin. Plasmin proteolytic activity digests fibrinogen and disrupts coagulation, exacerbated when α2 -antiplasmin is consumed by ongoing fibrinolysis. Additional direct inhibition of plasmin by aprotinin may prevent development of coagulopathy and extend the useful time window of TXA treatment.
Collapse
Affiliation(s)
- C. Longstaff
- Biotherapeutics DivisionNational Institute for Biological Standards and ControlSouth MimmsUK
| | - M. Locke
- Biotherapeutics DivisionNational Institute for Biological Standards and ControlSouth MimmsUK
| |
Collapse
|
138
|
Reversing anti–factor Xa agents and the unmet needs in trauma patients. Blood 2018; 132:2441-2445. [DOI: 10.1182/blood-2018-06-850396] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 10/09/2018] [Indexed: 12/14/2022] Open
Abstract
AbstractAndexanet alfa, a reversing agent for anticoagulants that inhibit factor Xa, has recently been licensed in the United States. We discuss the impact of this licensure on current practice and review in detail the problems of a neglected and growing clinical area: reversing the anticoagulation effect of factor Xa inhibitors in bleeding trauma patients. We identify areas of practice that need research so that care of bleeding trauma patients receiving direct factor Xa inhibitors can be improved.
Collapse
|
139
|
Shi W, Al-Sabti R, Burke PA, Gonzalez M, Mantilla-Rey N, Quillen K. Quality Management of massive transfusion protocol incorporating tranexamic acid adherence. Transfus Apher Sci 2018; 57:785-789. [PMID: 30455154 DOI: 10.1016/j.transci.2018.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 09/22/2018] [Accepted: 10/16/2018] [Indexed: 11/16/2022]
Abstract
Massive transfusion protocols (MTP) vary at different institutions. We implemented an algorithm in the transfusion service to support our Level I trauma center in 2007 and periodically monitor MTP utilization as part of ongoing quality management. At the last review in 2013, median plasma: RBC ratio was 1:1.8. We undertook a retrospective 3-year review of MTP activations stratifying by trauma versus non-trauma indications, and blood component utilization of the massive transfusion (MT) cases, adding a review of tranexamic acid (TXA) administration to the audit. The median transfused plasma: RBC ratio was 1:1.9 in trauma MT, and 1:1.6 in the non-trauma MT cases. Non-trauma MT patients at our institution were significantly older and more coagulopathic at MTP initiation compared to trauma MT patients, received fewer RBC units (15.5 versus 20.2), and had higher mortality. TXA adherence increased over the 3-year period to 60% of all trauma MTP activations in 2017.
Collapse
Affiliation(s)
- Weiwei Shi
- Department of Pathology and Laboratory Medicine, Boston University Medical Center, Boston, MA, 02118, United States.
| | - Ram Al-Sabti
- Department of Pathology and Laboratory Medicine, Boston University Medical Center, Boston, MA, 02118, United States
| | - Peter A Burke
- Department of Surgery, Boston University Medical Center, United States
| | - Mauricio Gonzalez
- Department of Anesthesiology, Boston University Medical Center, United States
| | - Nelson Mantilla-Rey
- Department of Pathology and Laboratory Medicine, Boston University Medical Center, Boston, MA, 02118, United States
| | - Karen Quillen
- Department of Pathology and Laboratory Medicine, Boston University Medical Center, Boston, MA, 02118, United States
| |
Collapse
|
140
|
Guillotte AR, Herbert JP, Madsen R, Hammer RD, Litofsky NS. Effects of platelet dysfunction and platelet transfusion on outcomes in traumatic brain injury patients. Brain Inj 2018; 32:1849-1857. [PMID: 30346865 DOI: 10.1080/02699052.2018.1536805] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
OBJECTIVE Platelet inhibition in traumatic brain injury (TBI) may be due to injury or antiplatelet medication use pre-injury. This study aims to identify factors associated with increased platelet arachidonic acid (AA) and adenosine diphosphate (ADP) inhibition and determine if platelet transfusion reduces platelet dysfunction and affects outcome. METHODS Prospective thromboelastography (TEG) assays were collected on adult patients with TBI with intracranial injuries detected by computed tomography (CT). Outcomes included in-hospital mortality, and CT lesion expansion. RESULTS Of 153 patients, ADP inhibition was increased in moderate and severe TBI compared to mild TBI (p = 0.0011). P2Y12 inhibiting medications had increased ADP inhibition (p = 0.0077). Admission ADP inhibition was not associated with in-hospital mortality (p = 0.24) or CT lesion expansion (p = 0.94). Mean reduction of ADP inhibition from platelet transfusion (-15.1%) relative to no transfusion (+ 11.7%) was not statistically different (p = 0.0472). CONCLUSIONS Mild TBI results in less ADP inhibition compared to moderate and severe TBI, suggesting a dose response relationship between TBI severity and degree of platelet dysfunction. Further, study is warranted to determine efficacy and parameters for platelet transfusion in patients with TBI.
Collapse
Affiliation(s)
- Andrew R Guillotte
- a Division of Neurological Surgery , University of Missouri School of Medicine , Columbia , MO , USA
| | - Joseph P Herbert
- a Division of Neurological Surgery , University of Missouri School of Medicine , Columbia , MO , USA
| | - Richard Madsen
- b Biostatistics and Research Design , University of Missouri School of Medicine , Columbia , MO , USA
| | - Richard D Hammer
- c Department of Pathology and Anatomical Sciences , University of Missouri School of Medicine , Columbia , MO USA
| | - N Scott Litofsky
- a Division of Neurological Surgery , University of Missouri School of Medicine , Columbia , MO , USA
| |
Collapse
|
141
|
Helin TA, Zuurveld M, Manninen M, Meijers JCM, Lassila R, Brinkman HJM. Hemostatic profile under fluid resuscitation during rivaroxaban anticoagulation: an in vitro survey. Transfusion 2018; 58:3014-3026. [DOI: 10.1111/trf.14933] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 08/13/2018] [Accepted: 08/15/2018] [Indexed: 12/27/2022]
Affiliation(s)
- Tuukka A. Helin
- Coagulation Disorders Unit, Clinical Chemistry; HUSLAB Laboratory Services, Helsinki University Hospital; Helsinki Finland
| | - Marleen Zuurveld
- Department of Molecular and Cellular Hemostasis; Sanquin Research; Amsterdam The Netherlands
| | | | - Joost C. M. Meijers
- Department of Molecular and Cellular Hemostasis; Sanquin Research; Amsterdam The Netherlands
- Amsterdam UMC, University of Amsterdam, Department of Experimental Vascular Medicine; Amsterdam Cardiovascular Sciences; Amsterdam The Netherlands
| | - Riitta Lassila
- Coagulation Disorders Unit, Clinical Chemistry; HUSLAB Laboratory Services, Helsinki University Hospital; Helsinki Finland
| | - Herm Jan M. Brinkman
- Department of Molecular and Cellular Hemostasis; Sanquin Research; Amsterdam The Netherlands
| |
Collapse
|
142
|
Early Detection and Management of Massive Intraoperative Pulmonary Embolism in a Patient Undergoing Repair of a Traumatic Acetabular Fracture. Case Rep Anesthesiol 2018; 2018:7485789. [PMID: 30364012 PMCID: PMC6188772 DOI: 10.1155/2018/7485789] [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] [Received: 06/05/2018] [Revised: 08/21/2018] [Accepted: 09/19/2018] [Indexed: 11/21/2022] Open
Abstract
A 73-year-old male with history of hyperlipidemia and osteoarthritis was transferred from an outside hospital after a fall from a ladder at home. He sustained a severe right sided acetabular fracture involving the femoral head, requiring operative repair. Preoperative evaluation was unremarkable except for oxygen saturation < 95 %. After induction of anesthesia and surgical positioning, the patient went into cardiac arrest. After intraoperative cardiopulmonary resuscitation (CPR) and placement on extracorporeal membrane oxygenation (ECMO), the patient stabilized. Cardiac catheterization revealed a large left pulmonary embolism. Here, we discuss the etiology and management of intraoperative pulmonary embolism.
Collapse
|
143
|
Abstract
Regional anesthesia for the acute trauma patient is increasing due to the growing appreciation of its benefits, development of newer techniques and equipment, and more robust training. Block procedures are expanding beyond perioperative interventions performed exclusively by anesthesiologists to paramedics on scene, emergency medicine physicians, and nurse-led services using these techniques early in trauma pain management. Special considerations and indications apply to trauma victims compared with the elective patient and must be appreciated to optimize safety and clinical outcomes. This review discusses current literature and future directions in the growing role of regional anesthesia in acute trauma care.
Collapse
Affiliation(s)
- Ian R Slade
- Department of Anesthesiology and Pain Medicine, Harborview Medical Center, University of Washington School of Medicine, 325 9th Avenue, Box 359724, Seattle, WA 98104, USA.
| | - Ron E Samet
- Department of Anesthesiology, Division of Trauma Anesthesiology, R Adams Cowley Shock Trauma Center, University of Maryland Medical Center, University of Maryland School of Medicine, 22 South Greene Street, Baltimore, MD 21201, USA
| |
Collapse
|
144
|
Rong H, Fan Y, Yang M, Zhang B, Sun D, Zhao Z, Wang D, Fan W, Wang J, Gu G, Li F, Liu X, Rao C, Chen H, Wang Y, Tian Y, Zhang J. Brain-derived microparticles activate microglia/macrophages and induce neuroinflammation. Brain Res 2018; 1694:104-110. [DOI: 10.1016/j.brainres.2018.05.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 05/02/2018] [Accepted: 05/15/2018] [Indexed: 12/13/2022]
|
145
|
Pohlman TH, Fecher AM, Arreola-Garcia C. Optimizing transfusion strategies in damage control resuscitation: current insights. J Blood Med 2018; 9:117-133. [PMID: 30154676 PMCID: PMC6108342 DOI: 10.2147/jbm.s165394] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
From clinical and laboratory studies of specific coagulation defects induced by injury, damage control resuscitation (DCR) emerged as the most effective management strategy for hemorrhagic shock. DCR of the trauma patient who has sustained massive blood loss consists of 1) hemorrhage control; 2) permissive hypotension; and 3) the prevention and correction of trauma-induced coagulopathies, referred to collectively here as acute coagulopathy of trauma (ACOT). Trauma patients with ACOT have higher transfusion requirements, may eventually require massive transfusion, and are at higher risk of exsanguinating. Distinct impairments in the hemostatic system associated with trauma include acquired quantitative and qualitative platelet defects, hypocoagulable and hypercoagulable states, and dysregulation of the fibrinolytic system giving rise to hyperfibrinolysis or a phenomenon referred to as fibrinolytic shutdown. Furthermore, ACOT is a component of a systemic host defense dysregulation syndrome that bears several phenotypic features comparable with other acute systemic physiological insults such as sepsis, myocardial infarction, and postcardiac arrest syndrome. Progress in the science of resuscitation has been continuing at an accelerated rate, and clinicians who manage catastrophic blood loss may be incompletely informed of important advances that pertain to DCR. Therefore, we review recent findings that further characterize the pathophysiology of ACOT and describe the application of this new information to optimization of resuscitation strategies for the patient in hemorrhagic shock.
Collapse
Affiliation(s)
- Timothy H Pohlman
- Department of Surgery, Lutheran Hospital of Indiana, Fort Wayne, IN, USA,
| | - Alison M Fecher
- Department of Surgery, Lutheran Hospital of Indiana, Fort Wayne, IN, USA
| | - Cecivon Arreola-Garcia
- Department of Surgery, Section of Acute Care Surgery, Indiana University Health, Indianapolis, IN, USA
| |
Collapse
|
146
|
Bouzat P, Ageron FX, Charbit J, Bobbia X, Deras P, Nugues JBD, Escudier E, Marcotte G, Leone M, David JS. Modelling the association between fibrinogen concentration on admission and mortality in patients with massive transfusion after severe trauma: an analysis of a large regional database. Scand J Trauma Resusc Emerg Med 2018; 26:55. [PMID: 29986757 PMCID: PMC6038237 DOI: 10.1186/s13049-018-0523-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 06/28/2018] [Indexed: 01/06/2023] Open
Abstract
Background The relationship between fibrinogen concentration and traumatic death has been poorly explored after severe trauma. Existing studies analysed this relationship in unselected trauma population, often considering fibrinogen concentration as a categorical variable. The aim of our study was to model the relationship between fibrinogen concentration and in-hospital mortality in severe trauma patients requiring massive transfusion using fibrinogen on admission as a continuous variable. Methods We designed a retrospective observational study based on prospectively collected data from 2009 to 2015 in seven French level-I trauma centres. All consecutive patients requiring a transfusion of at least 10 packed red blood cells (RBC) within 24 h were included. To assess the relationship between in-hospital death and fibrinogen concentration on admission, we performed generalized linear and additive models with death as a dependent variable. We also assessed the relationship between fibrinogen concentration below 1.5 g.L− 1 and potential predictors. Results Within the study period, 366 patients were included. A non-linear relationship was found between fibrinogen concentration and death. Graphical modelling of this relationship depicted a negative association between fibrinogen levels and death below a fibrinogen concentration of 1.5 g.L− 1. Predictors of low fibrinogen concentration (< 1.5 g.L− 1) were systolic blood pressure, Glasgow coma scale and haemoglobin concentration on admission. Conclusions A complex and robust approach for modelling the relationship between fibrinogen and mortality revealed a critical fibrinogen threshold of 1.5 g.L− 1 for severe trauma patients requiring massive transfusion. This trigger may guide the administration of procoagulant therapies in this context.
Collapse
Affiliation(s)
- Pierre Bouzat
- Grenoble Alps Trauma center, Department of anesthesiology and intensive care medicine, Grenoble University Hospital, F-38000, Grenoble, France. .,Grenoble Alps University, F-38000, Grenoble, France. .,Pôle d'Anesthésie-Réanimation, Hôpital Albert Michallon, 217, F-38043, Grenoble, BP, France.
| | - François-Xavier Ageron
- RENAU Northern French Alps Emergency Network, Public Health department, Annecy Hospital, F-74000, Annecy, France.,Department of emergency medicine and intensive care, Annecy Hospital, F-74000, Annecy, France
| | - Jonathan Charbit
- Department of anesthesiology and intensive care, Montpellier University Hospital, F-34000, Montpellier, France
| | - Xavier Bobbia
- Department of emergency medicine, Nimes University Hospital, F-30000, Nimes, France
| | - Pauline Deras
- Department of anesthesiology and intensive care, Montpellier University Hospital, F-34000, Montpellier, France
| | - Jennifer Bas Dit Nugues
- Grenoble Alps Trauma center, Department of anesthesiology and intensive care medicine, Grenoble University Hospital, F-38000, Grenoble, France
| | - Etienne Escudier
- Department of emergency medicine and intensive care, Annecy Hospital, F-74000, Annecy, France
| | - Guillaume Marcotte
- Department of anesthesiology and intensive care, Lyon-Edouard Herriot University Hospital, F-69000, Lyon, France
| | - Marc Leone
- Aix Marseille University, Nord Hospital, Department of anesthesiology and intensive medicine, APHM, F-13000, Marseille, France
| | - Jean-Stéphane David
- Hospices Civils de Lyon, Lyon-Sud University Hospital, Department of anesthesiology and intensive care, F-69495, Pierre-Bénite, France.,Claude Bernard Lyon 1 University, F-69008, Lyon, France
| |
Collapse
|
147
|
Barelli S, Alberio L. The Role of Plasma Transfusion in Massive Bleeding: Protecting the Endothelial Glycocalyx? Front Med (Lausanne) 2018; 5:91. [PMID: 29721496 PMCID: PMC5915488 DOI: 10.3389/fmed.2018.00091] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Accepted: 03/22/2018] [Indexed: 12/20/2022] Open
Abstract
Massive hemorrhage is a leading cause of death worldwide. During the last decade several retrospective and some prospective clinical studies have suggested a beneficial effect of early plasma-based resuscitation on survival in trauma patients. The underlying mechanisms are unknown but appear to involve the ability of plasma to preserve the endothelial glycocalyx. In this mini-review, we summarize current knowledge on glycocalyx structure and function, and present data describing the impact of hemorrhagic shock and resuscitation fluids on glycocalyx. Animal studies show that hemorrhagic shock leads to glycocalyx shedding, endothelial inflammatory changes, and vascular hyper-permeability. In these animal models, plasma administration preserves glycocalyx integrity and functions better than resuscitation with crystalloids or colloids. In addition, we briefly present data on the possible plasma components responsible for these effects. The endothelial glycocalyx is increasingly recognized as a critical component for the physiological vasculo-endothelial function, which is destroyed in hemorrhagic shock. Interventions for preserving an intact glycocalyx shall improve survival of trauma patients.
Collapse
Affiliation(s)
- Stefano Barelli
- Division of Haematology and Central Haematology Laboratory, CHUV, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Lorenzo Alberio
- Division of Haematology and Central Haematology Laboratory, CHUV, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland.,Faculté de Biologie et Médecine, UNIL, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
148
|
Is Coagulopathy an Appropriate Therapeutic Target During Critical Illness Such as Trauma or Sepsis? Shock 2018; 48:159-167. [PMID: 28234791 DOI: 10.1097/shk.0000000000000854] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Coagulopathy is a common and vexing clinical problem in critically ill patients. Recently, major advances focused on the treatment of coagulopathy in trauma and sepsis have emerged. However, the targeting of coagulopathy with blood product transfusion and drugs directed at attenuating the physiologic response to these conditions has major potential risk to the patient. Therefore, the identification of coagulopathy as a clinical target is an area of uncertainty and controversy. To analyze the state of the science regarding coagulopathy in critical illness, a symposium addressing the problem was organized at the 39th annual meeting of the Shock Society in the summer of 2016. This manuscript synthesizes the viewpoints of the four expert panelists at the debate and presents an overview of the potential positive and negative consequences of targeting coagulopathy in trauma and sepsis.
Collapse
|
149
|
Halbgebauer R, Braun CK, Denk S, Mayer B, Cinelli P, Radermacher P, Wanner GA, Simmen HP, Gebhard F, Rittirsch D, Huber-Lang M. Hemorrhagic shock drives glycocalyx, barrier and organ dysfunction early after polytrauma. J Crit Care 2018; 44:229-237. [DOI: 10.1016/j.jcrc.2017.11.025] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 10/30/2017] [Accepted: 11/16/2017] [Indexed: 10/18/2022]
|
150
|
Holcomb JB, Hess JR. Response to: "Misunderstanding the PROPPR trial". Transfusion 2018; 57:2057-2058. [PMID: 28782821 DOI: 10.1111/trf.14197] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 05/10/2017] [Indexed: 12/30/2022]
Affiliation(s)
- John B Holcomb
- Professor of Surgery, Center for Translational Injury Research, University of Texas Health Science Center, Houston, Texas
| | - John R Hess
- Harborview Transfusion Service, Harborview Medical Center, Seattle, Washington
| | | |
Collapse
|