Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
Add more filters










Publication year range
1.
Surgery ; 175(3): 613-617, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37863690

ABSTRACT

BACKGROUND: The endothelial glycocalyx is a critical component of the vascular barrier; its disruption after shock states may contribute to coagulopathy in a variety of conditions. Measurement of glycocalyx components in plasma have been used to index glycocalyx degradation but are not available as a point of care test. Heparanoids, such as heparan sulfate, may affect coagulation which may be detected by either thromboelastography or activated clotting time. METHODS: Endothelial glycocalyx components syndecan-1 and heparan sulfate were added to blood samples at clinically relevant concentrations. Thromboelastography values included clot reaction time, clot amplification and fibrinogen values, and maximum clot strength (maximum amplitude, platelets). The heparinase thromboelastography cartridge was used to detect a heparin-like effect. The activated clotting time test was performed subsequently using the heparan sulfate blood samples to compare a standard coagulation test with thromboelastography clot reaction times. RESULTS: Both thromboelastography clot reaction time (with comparison to heparinase) and activated clotting time were useful to detect effects of coagulation. Thromboelastography also detected platelet and fibrinogen abnormalities at higher heparan sulfate concentrations. Studies using thromboelastography or even activated clotting time may be useful to detect glycocalyx degradation after shock states and may guide clinical decision making. CONCLUSION: Thromboelastography and or activated clotting time may be useful to detect glycocalyx degradation as a point of care test in patients in the acute setting. Additionally, these assays may detect previous undisclosed coagulopathy due to glycocalyx degradation.


Subject(s)
Blood Coagulation Disorders , Thrombelastography , Humans , Glycocalyx/metabolism , Heparin Lyase/metabolism , Blood Coagulation Disorders/diagnosis , Blood Coagulation Disorders/etiology , Fibrinogen , Heparitin Sulfate/metabolism
2.
J Trauma Acute Care Surg ; 94(2): 273-280, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36322025

ABSTRACT

BACKGROUND: Traumatic/hemorrhagic shock, sepsis and other inflammatory processes lead to endothelial activation and a loss of the endothelial glycocalyx. von Willebrand factor (vWF) is an acute phase reactant that is released from endothelial cells and megakaryocytes. Stimulated but not basal vWF leads to significant formation of ultralarge multimers (ultralarge vWF [ULvWF]) and risk for thrombotic complications. Ultralarge vWF is cleaved by a disintegrin and metalloproteinase with a thrombospondin type motif 13 (ADAMTS 13); alterations in ULvWF/ADAMTS 13 ratio may contribute to trauma-induced coagulopathy. Salutary effects of tranexamic acid (TXA) on trauma-induced coagulopathy have been described. These effects appear apart from antifibrinolytic actions of TXA and include protection of the endothelial glycocalyx. Ultralarge vWF is in part anchored to the glycocalyx layer of the endothelium. Tranexamic acid protected the endothelial glycocalyx layer from degradation using a microfluidic model of the microcirculation subjected to hypoxia-reoxygenation and catecholamine excess. We hypothesized that TXA administration following shock conditions would impact the vWF-ADAMTS-13 axis by protecting the glycocalyx from degradation. This was studied in a endothelial microfluidic flow study. METHODS: Human umbilical vein endothelial cells were established under flow conditions and subjected to biomimetic shock. Tranexamic acid was added after 90 minutes of perfusion. von Willebrand factor antigen and ADAMTS-13 activity were measured. Western blot analysis was performed for vWF characterization from perfusion media. RESULTS: Shock conditions increased vWF antigen and decreased ADAMTS 13 activity. Tranexamic acid ameliorated shock induced cleavage in the ADAMTS 13-vWF axis with a reduction of the thrombogenic ULvWF. CONCLUSION: These results suggest another mechanism whereby administration of TXA early following traumatic/hemorrhagic shock mitigates microvascular perfusion abnormalities and subsequent organ failure. The resultant effects on platelet adhesion and aggregation require further study.


Subject(s)
Shock, Hemorrhagic , Tranexamic Acid , Humans , von Willebrand Factor/metabolism , Tranexamic Acid/pharmacology , Tranexamic Acid/therapeutic use , ADAM Proteins/metabolism , ADAMTS13 Protein/metabolism , Shock, Hemorrhagic/metabolism , Endothelium, Vascular/metabolism , Human Umbilical Vein Endothelial Cells/metabolism
3.
Surgery ; 173(3): 876-882, 2023 03.
Article in English | MEDLINE | ID: mdl-36372576

ABSTRACT

BACKGROUND: Studies have suggested a beneficial effect of early plasma-based resuscitation in patients following trauma-hemorrhagic shock. The underlying mechanism(s) are unknown but may be owing to protective effects of plasma components on the endothelium and its glycocalyx layer. Albumin, the major protein in plasma, influences vascular integrity and has antioxidant properties in vivo. Sphingosine 1-phosphate is a bioactive sphingolipid with diverse signaling functions, which include endothelial barrier protection in part owing to preservation of the glycocalyx. Sphingosine 1-phosphate is bound mainly to albumin and high-density lipids in the plasma. Debate continues about the beneficial effect of albumin solutions in shock resuscitation. Pharmacologic preparations may modify constituents of albumin solutions for clinical use. We examined the relative effects of sphingosine 1-phosphate concentrations in albumin solutions on the endothelial-glycocalyx barrier in an in vitro microfluidic platform. METHODS: Endothelial cell monolayers were established in microfluidic perfusion devices and exposed to control or biomimetic shock conditions followed by 5% plasma or different albumin solutions ± exogenous sphingosine 1-phosphate perfusion. Biomarkers of endothelial and glycocalyx activation, damage, and oxidant injury were then determined. RESULTS: Endothelial cell and glycocalyx barriers were damaged after biomimetic shock conditions. Plasma and sphingosine 1-phosphate loaded albumin solutions protected against barrier injury. Modest protective effects were noted with albumin alone; the efficacy varied with sphingosine 1-phosphate content of the albumin solution. CONCLUSION: The protective effect of albumin on the endothelia-glycocalyx barrier against oxidant injury was dependent on its sphingosine 1-phosphate concentration. Our data may help explain the discrepancies regarding the effectiveness of albumin solutions in shock resuscitation.


Subject(s)
Microfluidics , Oxidants , Humans , Oxidants/metabolism , Endothelium/metabolism , Albumins
4.
J Trauma Acute Care Surg ; 92(5): 812-820, 2022 05 01.
Article in English | MEDLINE | ID: mdl-35067525

ABSTRACT

BACKGROUND: Improved outcomes with early tranexamic acid (TXA) following trauma hemorrhagic shock (T/HS) may be related to its antifibrinolytic, as well as anti-inflammatory properties. Previous in vitro studies have shown that early TXA administration protects against T/HS endothelial barrier dysfunction and associated glycocalyx degradation. An intact endothelial glycocalyx may protect against subsequent neutrophil mediated tissue injury. We postulated that early TXA administration would mitigate against glycocalyx damage and resultant neutrophil adherence and transmigration through the endothelial barrier. This was studied in vitro using a microfluidic flow platform. METHODS: Human umbilical vein endothelial cell monolayers were subjected to control or shock conditions (hypoxia + epinephrine) followed by administration of TXA 90 minutes or 180 minutes later. RESULTS: "Early" TXA administration protected against glycocalyx degradation, biomarkers of increased permeability and the development of a fibrinolytic phenotype. This was associated with decreased neutrophil endothelial adherence and transmigration. There were no differences in low versus high TXA concentrations. The protective effects were only significant with "early" TXA administration. CONCLUSION: There was a concentration and temporal effect of TXA administration on endothelial glycocalyx degradation. This was associated with "vascular leakiness" as indexed by the relative ratio of Ang-2/1 and polymorphonuclear neutrophil transmigration. Tranexamic acid if administered in patients with T/HS should be administered "early"; this includes in the prehospital setting.


Subject(s)
Antifibrinolytic Agents , Shock, Hemorrhagic , Tranexamic Acid , Antifibrinolytic Agents/pharmacology , Glycocalyx/metabolism , Humans , Permeability , Tranexamic Acid/pharmacology , Tranexamic Acid/therapeutic use
5.
Surgery ; 171(3): 825-832, 2022 03.
Article in English | MEDLINE | ID: mdl-34865862

ABSTRACT

BACKGROUND: Hemorrhagic shock leads to endothelial glycocalyx shedding, endothelial cellular inflammation, and increased vascular permeability. Early plasma administration improves survival in severely injured patients; this may be due in part to its ability to ameliorate this trauma-induced endotheliopathy. The protective effect of early plasma administration may be due to its sphingosine 1-phosphate content. Principle carriers of plasma sphingosine 1-phosphate include apolipoprotein M and albumin. The relative roles of these carriers on sphingosine 1-phosphate protective effects are unknown and were studied in an in vitro model of microcirculation. METHODS: Endothelial cell monolayers were established in microfluidic perfusion devices and exposed to control or biomimetic shock conditions. Sphingosine 1-phosphate, albumin + sphingosine 1-phosphate, or apolipoprotein M + sphingosine 1-phosphate were added later to the perfusate. Biomarkers of endothelial and glycocalyx activation and damage were then determined. RESULTS: Sphingosine 1-phosphate preserved endothelial and glycocalyx barrier function after exposure to conditions of shock in the microcirculation. The protective effect was related to sphingosine 1-phosphate chaperones; the apolipoprotein M loaded with sphingosine 1-phosphate had the most profound effect. CONCLUSION: Carrier-based sphingosine 1-phosphate may be a useful adjunct in early hemorrhagic shock resuscitation.


Subject(s)
Human Umbilical Vein Endothelial Cells/drug effects , Lysophospholipids/pharmacology , Shock/pathology , Sphingosine/analogs & derivatives , Albumins/pharmacology , Apolipoproteins M/pharmacology , Cell Culture Techniques , Glycocalyx/drug effects , Humans , Microcirculation , Sphingosine/pharmacology
6.
J Trauma Acute Care Surg ; 91(5): 849-855, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34695061

ABSTRACT

BACKGROUND: Aging is characterized by a decline in cellular function, which has an adverse effect on the biologic response to injury. Both aging and trauma/hemorrhagic shock (T/HS) increase oxidative stress which impairs the vascular endothelium (EC) and glycocalyx (EG). The additive effect of aging on EC and EG damage following T/HS are unknown. This was studied in an in vitro model. METHODS: Confluent endothelial cell monolayers from primary aortic endothelial cells from 10-week-old mice ("young" cells) or primary aortic cells from 65-week-old mice ("aged" cells) were established in microfluidic devices (MFDs) and perfused at constant shear conditions overnight. Mouse endothelial cell monolayers were then exposed to hypoxia/reoxygenation alone and/or epinephrine or norepinephrine. Endothelial glycocalyx degradation was indexed as well as subsequent endothelial injury/activation. RESULTS: Aged endothelial cells showed increase glycocalyx shedding and subsequent loss of glycocalyx thickness. This lead to a more pronounced level of EC injury/activation compared with young endothelial cells. Although exposure to biomimetic shock conditions exacerbated both endothelial glycocalyx shedding and endothelial injury in both aged and young endothelial cells, the effect was significantly more pronounced in aged cells. CONCLUSION: Advanced age is associated with worse outcomes in severely injured trauma patients. Our study demonstrates that there is increased EG shedding and a diminished EG layer in aged compared to "young" endothelial cell layers. Biomimetic shock conditions lead to an even greater impairment of the endothelial glycocalyx in aged versus young endothelial cell monolayers. It appears that these effects are a consequence of aging related oxidative stress at both baseline and shock conditions. This exacerbates shock-induced endotheliopathy and may contribute to untoward effects on patient outcomes in this population.


Subject(s)
Aging/physiology , Endothelium, Vascular/physiopathology , Glycocalyx/physiology , Shock, Hemorrhagic/physiopathology , Aged , Animals , Biomimetic Materials , Cells, Cultured , Disease Models, Animal , Humans , Mice , Microfluidic Analytical Techniques , Oxidative Stress/physiology , Reactive Oxygen Species/metabolism , Young Adult
7.
Surgery ; 169(3): 678-685, 2021 03.
Article in English | MEDLINE | ID: mdl-32988619

ABSTRACT

BACKGROUND: Sexual dimorphism has been demonstrated after major trauma and hemorrhage shock with protective effects related to female sex or estrogen. Traumatic endotheliopathy is an important component of trauma-induced coagulopathy. Components of endothelial barrier dysfunction include degradation of the endothelial glycocalyx and endothelial cellular injury. Estrogen modulates endothelial function via its membrane and cellular receptors. The effects of estrogen on the vascular endothelial barrier after trauma and hemorrhage shock are, however, unknown. This topic was studied in an in vitro model under flow conditions. METHODS: Monolayers of human umbilical vein endothelial cells were established in microfluidic flow devices. After overnight perfusion, cell monolayers were subjected to normoxic or hypoxic perfusion and then treated with either estrogen (as estradiol), testosterone (as dihydrotestosterone), or media alone. Endothelial activation/injury was indexed by soluble thrombomodulin and glycocalyx degradation by syndecan-1 and hyaluronic acid shedding as well as measurement of the thickness of the glycocalyx layer. The coagulation phenotype of the human umbilical vein endothelial cells was indexed by the relative values of the activities of tissue plasminogen activator and plasminogen activator inhibitor-1. Vascular endothelial growth factor was measured in cell culture supernatants using a solid-phase enzyme-linked immunosorbent assay. RESULTS: Treatment with estrogen but not testosterone mitigated the adverse effect of shock on endothelial and glycocalyx barrier properties. Our biomimetic model suggests a beneficial effect of estrogen administration after trauma and hemorrhage shock on the glycocalyx and endothelial barriers. CONCLUSION: Early estrogen treatment after trauma and hemorrhage shock may be a useful adjunct to mitigating the development of traumatic endotheliopathy.


Subject(s)
Endothelial Cells/metabolism , Estrogens/metabolism , Glycocalyx/metabolism , Protective Agents/metabolism , Shock/etiology , Shock/metabolism , Biomarkers , Cells, Cultured , Endothelial Cells/drug effects , Estrogens/pharmacology , Female , Fluorescent Antibody Technique , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Male , Microfluidics/methods , Phenotype , Protective Agents/pharmacology , Proteinase Inhibitory Proteins, Secretory/metabolism , Sex Factors , Vascular Endothelial Growth Factor A/metabolism
8.
J Trauma Acute Care Surg ; 89(3): 544-550, 2020 09.
Article in English | MEDLINE | ID: mdl-32467464

ABSTRACT

BACKGROUND: Impaired microvascular perfusion in the obese patient has been linked to chronic adverse health consequences. The impact on acute illnesses including trauma, sepsis, and hemorrhagic shock (HS) is uncertain. Studies have shown that endothelial glycocalyx and vascular endothelial derangements are causally linked to perfusion abnormalities. Trauma and HS are also associated with impaired microvascular perfusion in which glycocalyx injury and endothelial dysfunction are sentinel events. We postulate that obesity may impact the adverse consequences of HS on the vascular barrier. This was studied in vivo in a biomimetic model of HS using microfluidic technology. METHODS: Human umbilical vein endothelial cell monolayers were established in a microfluidic device. Cells were exposed to standard or biomimetic shock conditions (hypoxia plus epinephrine) followed by perfusion from plasma obtained from obese or nonobese subjects. Endothelial glycocalyx and endothelial cellular injury were then determined. RESULTS: Plasma from nonobese patients completely reversed glycocalyx and endothelial vascular barrier injury. Plasma from obese patients was only partially protective and was associated with differences in adipokines and other substances in the plasma of these patients. CONCLUSION: Our study supports that obesity impairs HS resuscitation. This may be due to microrheological differences between nonobese and obese individuals and may contribute to the poorer outcome in this patient population.


Subject(s)
Endothelium, Vascular/physiopathology , Microfluidics/methods , Obesity/physiopathology , Plasma , Shock, Hemorrhagic/therapy , Wounds and Injuries/complications , Biomimetics , Cells, Cultured , Female , Glycocalyx , Human Umbilical Vein Endothelial Cells/physiology , Humans , Lab-On-A-Chip Devices , Male , Resuscitation/methods , Shock, Hemorrhagic/etiology , Shock, Hemorrhagic/physiopathology , Vasodilation/physiology , Wounds and Injuries/physiopathology
9.
J Trauma Acute Care Surg ; 87(5): 1061-1069, 2019 11.
Article in English | MEDLINE | ID: mdl-31453986

ABSTRACT

BACKGROUND: Plasma is an important component of resuscitation after trauma and hemorrhagic shock (T/HS). The specific plasma proteins and the impact of storage conditions are uncertain. Utilizing a microfluidic device system, we studied the effect of various types of plasma on the endothelial barrier function following T/HS. METHODS: Human umbilical vein endothelial cells (HUVEC) were cultured in microfluidic plates. The microfluidic plates were subjected to control or shock conditions (hypoxia/reoxygenation + epinephrine, 10 µM). Fresh plasma, 1 day thawed plasma, 5-day thawed plasma and lyophilized plasma were then added. Supplementation of sphingosine-1 phosphate (S-1P) was done in a subset of experiments. Effect on the endothelial glycocalyx was indexed by shedding of syndecan-1 and hyaluronic acid. Endothelial injury/activation was indexed by soluble thrombomodulin, tissue plasminogen activator, plasminogen activator inhibitor-1. Vascular permeability determined by the ratio of angiopoietin-2 to angiopoietin-1. Concentration of S-1P and adiponectin in the different plasma groups was measured. RESULTS: Human umbilical vein endothelial cells exposed to shock conditions increased shedding of syndecan-1 and hyaluronic acid. Administration of the various types of plasma decreased shedding, except for 5-day thawed plasma. Shocked HUVEC cells demonstrated a profibrinolytic phenotype, this normalized with all plasma types except for 5-day thawed plasma. The concentration of S-1P was significantly less in the 5-day thawed plasma compared with the other plasma types. Addition of S-1P to 5-day thawed plasma returned the benefits lost with storage. CONCLUSION: A biomimetic model of the microcirculation following T/HS demonstrated endothelial glycocalyx and endothelial cellular injury/activation as well as a profibrinolytic phenotype. These effects were abrogated by all plasma products except the 5-day thawed plasma. Plasma thawed longer than 5 days had diminished S1-P concentrations. Our data suggest that S1-P protein is critical to the protective effect of plasma products on the endothelial-glycocalyx barrier following T/HS.


Subject(s)
Blood Component Transfusion/methods , Endothelium/metabolism , Plasma/metabolism , Shock, Hemorrhagic/therapy , Wounds and Injuries/therapy , Cell Line , Endothelium/cytology , Endothelium/pathology , Glycocalyx/metabolism , Glycocalyx/pathology , Human Umbilical Vein Endothelial Cells , Humans , Lysophospholipids/metabolism , Microfluidic Analytical Techniques , Resuscitation/methods , Shock, Hemorrhagic/etiology , Shock, Hemorrhagic/pathology , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Wounds and Injuries/complications
10.
J Trauma Acute Care Surg ; 86(6): 943-951, 2019 06.
Article in English | MEDLINE | ID: mdl-31124891

ABSTRACT

BACKGROUND: Observational studies have identified an association between duration of red blood cell (RBC) storage and adverse outcomes in trauma. Hemorrhagic shock (HS) leads to impaired tissue perfusion which is associated with endothelial cell glycocalyx (eGC) shedding. Adhesion of stored RBC to the vascular endothelium has been shown to lead to impaired perfusion in the microcirculation and contribute to organ failure and poor outcome. The role of either or both of the EC and RBC glycocalyx in this process is unknown and was studied in an in vitro model. METHODS: Human umbilical vein endothelial cells were perfused in a microfluidic device with RBC solutions from fresh, less than 14-day or longer than 21-day storage. In some experiments, the HS microenvironment was simulated by hypoxia-reoxygenation (H/R) and epinephrine (Epi) in the perfusion experiments. Measurements obtained included endothelial cell (EC) and RBC glycocalyx and RBC adherence to human umbilical vein endothelial cell monolayers at variable shear rates. RESULTS: Endothelial cell glycocalyx and RBC glycocalyx dimensions were reduced by H/R and Epi and storage duration respectively. Red blood cell adherence to the endothelium was increased by H/R + Epi treatment and duration of RBC storage. CONCLUSION: Our data may help explain some of the remaining discrepancies regarding the impact of RBC storage duration on outcomes in the trauma population. Consideration of the integrity of the EC and RBC glycocalyx may guide future transfusion strategies in the trauma population. The microfluidic device system platform may offer a high throughput modality to study emerging therapies to mitigate adverse consequence of RBC storage duration on the perfused endothelium in the trauma setting.


Subject(s)
Cell Adhesion/physiology , Endothelium, Vascular/physiology , Erythrocytes/physiology , Microcirculation/physiology , Shock, Hemorrhagic/physiopathology , Stress, Physiological/physiology , Blood Preservation/methods , Glycocalyx/metabolism , Humans , Microfluidics/methods
11.
Am J Surg ; 217(6): 1076-1082, 2019 06.
Article in English | MEDLINE | ID: mdl-30635208

ABSTRACT

BACKGROUND: Hyperglycemia promotes vascular inflammation; however its effect on endothelial dysfunction in sepsis is unknown. Microfluidic devices (MFD) may closely mimic the in vivo endothelial cell microenvironment. We hypothesized that stress glucose concentrations would increase sepsis related endothelial injury/activation. METHODS: Human umbilical vein endothelial cell (HUVEC) monolayers were established in microfluidic channels. TNF was added followed by glucose. Endothelial glycocalyx (EG) integrity was indexed by shedding of the EG components as well as thickness. Endothelial cell (EC) injury/activation was indexed by soluble biomarkers. Intracellular reactive oxygen species (ROS) was by fluorescence. RESULTS: TNF increased glycocalyx degradation and was associated with biomarkers of EC injury. These vascular barrier derangements were further increased by hyperglycemia. This may be related to increase ROS species generated followed by the combined insults. CONCLUSION: MFD technology may be a useful platform to study endothelial barrier function and stress conditions and allow preclinical assessment of potential therapies.


Subject(s)
Glycocalyx/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Hyperglycemia/metabolism , Microfluidics , Sepsis/metabolism , Acute Disease , Biomarkers/metabolism , Humans , Optical Imaging , Reactive Oxygen Species/metabolism , Stress, Physiological/physiology , Tumor Necrosis Factor-alpha/metabolism
12.
J Surg Res ; 232: 195-201, 2018 12.
Article in English | MEDLINE | ID: mdl-30463718

ABSTRACT

BACKGROUND: Clostridium difficile infection (CDI) is due to the effects of toxins, toxin A and toxin B on the host. Severe CDI is associated with systemic signs of infection. Animal models of CDI demonstrate a strong correlation between systemic toxemia and the occurrence of severe disease. However, current technologies have low sensitivity to detect C difficile toxemia in human subjects. Raman spectroscopy (RS) is an upcoming technology that is used to detect bacteria and their toxins. We speculate that RS may be a sensitive method to detect clinically relevant concentrations of C difficile toxins in serum. MATERIALS AND METHODS: Serum samples were spiked with varying concentrations of toxin A, toxin B, and both. RS was performed on an air-dried serum drop that was placed on a mirror-polished stainless steel slide. Raman spectra were obtained, background corrected, vector normalized, and analyzed by Partial Least Square Linear Discriminant Analysis and Support Vector Machine for Classification. Model accuracy was measured by cross-validation and bootstrap methods. RESULTS: Toxin-spiked sera of various concentrations (1 ng/mL, 1 pg/mL, and 0.1 pg/mL) were distinguished from control serum 100% with cross-validation error rate ranging from 0% to 18% and bootstrap error rate ranging from 0% to 12% for various concentrations. The sensitivity ranged from 87% to 100% and specificity ranged from 77% to 100% for various concentrations of toxin-spiked serum. CONCLUSIONS: We conclude that RS may be a sensitive method to detect clinically relevant concentrations of C difficile toxins in serum and thus to help diagnose severe CDI in patients in real-time at the point of care.


Subject(s)
Bacterial Proteins/blood , Bacterial Toxins/blood , Enterotoxins/blood , Spectrum Analysis, Raman/methods , Humans , Least-Squares Analysis
13.
J Trauma Acute Care Surg ; 85(5): 960-967, 2018 11.
Article in English | MEDLINE | ID: mdl-29851906

ABSTRACT

BACKGROUND: Early hyperglycemia is associated with higher mortality in trauma and predicts multiple organ failure. Endothelial cell (EC) injury and glycocalyx (GC) degradation occur following traumatic shock and are key factors in the development of trauma-induced coagulopathy and result in impaired microvascular perfusion and accompanying organ failure. Acute hyperglycemia has been shown to result in the loss of the GC layer, EC inflammation, and activation of coagulation in vivo. We postulated that acute hyperglycemia would exacerbate trauma-induced EC injury and GC shedding and integrity. This was studied using a microfluidic device in a biomimetic in vitro model. METHODS: Human umbilical vein endothelial cell monolayers established in the microfluidic channels of a microfluidic device well plate were perfused at constant shear overnight. Human umbilical vein endothelial cell monolayers were then exposed to hypoxia/reoxygenation and epinephrine followed by the addition of varying concentrations of glucose. RESULTS: Glycocalyx shedding and loss of dimension, as well as EC injury/activation, were noted after exposure to the biomimetic conditions of trauma/shock in our study. Similar but less dramatic findings were noted after acute hyperglycemia. Exposure to hyperglycemia exacerbated the adverse effects on the GC and EC following hypoxia/reoxygenation plus epinephrine exposure and may be related to enhanced production of reactive oxygen species. CONCLUSIONS: Microfluidic device study may allow the preclinical assessment and development of therapeutic strategies of the vascular barrier under stress conditions.


Subject(s)
Glucose/pharmacology , Glycocalyx/pathology , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Acute Disease , Adrenergic beta-Agonists/pharmacology , Angiopoietin-1/metabolism , Angiopoietin-2/metabolism , Cell Hypoxia , Cells, Cultured , Epinephrine/pharmacology , Humans , Hyaluronic Acid/metabolism , Hyperglycemia/complications , In Vitro Techniques , Lab-On-A-Chip Devices , Oxygen/administration & dosage , Reactive Oxygen Species/metabolism , Syndecan-1/metabolism , Wounds and Injuries/blood , Wounds and Injuries/pathology
14.
J Trauma Acute Care Surg ; 85(1): 128-134, 2018 07.
Article in English | MEDLINE | ID: mdl-29538236

ABSTRACT

BACKGROUND: Hypernatremia is a common problem affecting critically ill patients, whether due to underlying pathology or the subsequent result of hypertonic fluid resuscitation. Numerous studies have been published, suggesting that hypernatremia may adversely affect the vascular endothelial glycocalyx. Our study aimed to evaluate if high sodium concentration would impair the endothelial and glycocalyx barrier function and if stress conditions that simulate the shock microenvironment would exacerbate any observed adverse effects of hypernatremia. METHODS: Human umbilical vein endothelial cells (HUVEC) were cultured in microfluidic channels subjected to flow conditions overnight to stimulate glycocalyx growth. Cells were then subjected to sodium (Na) concentrations of either 150 mEq/L or 160 mEq/L, with Hepes solution applied to media to maintain physiologic pH. Subsets of HUVEC were also exposed to hypoxia/reoxygenation and epinephrine (HR + Epi) to simulate shock insult, then followed by Na treatment. Perfusate was then collected 60 minutes and 120 minutes following treatments. Relevant biomarkers were then evaluated and HUVEC underwent fluorescent staining followed by microscopy. RESULTS: Glycocalyx degradation as indexed by hyaluronic acid and syndecan-1 was elevated in all subgroups, particularly those subjected to HR + Epi with Na 160 mEq/L. Thickness of the glycocalyx as evaluated by fluorescent microscopy was reduced to half of baseline with Na 160 mEq/L and to one third of baseline with additional insult of HR + Epi. Endothelial activation/injury as indexed by soluble thrombomodulin was elevated in all subgroups. A profibrinolytic coagulopathy phenotype was demonstrated in all subgroups with increased tissue plasminogen activator levels and decreased plasminogen activator inhibitor-1 levels. CONCLUSION: Our data suggest that hypernatremia results in degradation of the endothelial glycocalyx with further exacerbation by shock conditions. A clinical study using clinical measurements of the endothelial glycocalyx in critically ill or injured patients with acquired hypernatremia would be warranted.


Subject(s)
Endothelium, Vascular/physiopathology , Glycocalyx/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Hypernatremia/physiopathology , Sodium/pharmacology , Biomarkers/metabolism , Cell Culture Techniques , Endothelium, Vascular/metabolism , Epinephrine/pharmacology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hypernatremia/complications , Microfluidics/methods , Plasminogen Activator Inhibitor 1/metabolism , Shock, Hemorrhagic/physiopathology , Syndecan-1/metabolism , Tissue Plasminogen Activator/metabolism
15.
J Trauma Acute Care Surg ; 84(1): 75-80, 2018 01.
Article in English | MEDLINE | ID: mdl-29040203

ABSTRACT

BACKGROUND: The endothelial glycocalyx (GCX) plays an important role in vascular barrier function. Damage to the GCX occurs due to a variety of causes including hypoxia, ischemia-reperfusion, stress-related sympathoadrenal activation, and inflammation. Tranexamic acid (TXA) may prevent GCX degradation. The therapeutic window for TXA administration and the mechanism of action has been under review. Membrane-anchored proteases (sheddases) are key components in endothelial cell biology including the regulation of vascular permeability. The effect of TXA administration on stress-related GCX damage, and the role of sheddases in this process was studied in a cell-based model. METHODS: Confluent human umbilical vein endothelial cells (HUVEC) were exposed to hydrogen peroxide and/or epinephrine (EPI) to stimulate postshock reperfusion. TXA was added at various times after hydrogen peroxide (H2O2) and/or EPI exposure. GCX degradation was indexed by syndecan-1 and hyaluronic acid release. Activation of endothelial sheddases was indexed by A Disintegrin and Metalloproteinase-17 and matrix metalloproteinase-9 activity in culture supernatants. RESULTS: Exposure of HUVEC to either/both EPI and H2O2 resulted in a cellular stress and GCX disruption demonstrated by increased levels of syndecan-1 shedding, hyaluronic acid release, tumor necrosis factor-α release. Shedding of these GCX components was associated with increased activity of both A Disintegrin and Metalloproteinase-17 and matrix metalloproteinase. Disruption of the GCX was further demonstrated via fluorescent imaging, which demonstrated disruption after exposure to either/both H2O2 and EPI. Early administration of either TXA or doxycycline resulted in preservation of the GCX. Late administration of TXA had no effect, whereas doxycycline had some residual protective effect. CONCLUSION: Tranexamic acid as a serine protease inhibitor prevented GCX degradation via inhibition of endothelial sheddase activation. This effect was not apparent when TXA was administered greater than 60 minutes after "simulated" reperfusion. Our study supports the clinical practice of early TXA administration in the severely injured patient.


Subject(s)
Doxycycline/pharmacology , Glycocalyx/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Protease Inhibitors/pharmacology , Tranexamic Acid/pharmacology , ADAM17 Protein/metabolism , Cell Culture Techniques , Epinephrine , Glycocalyx/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hyaluronic Acid , Hydrogen Peroxide , Oxidative Stress , Syndecan-1/metabolism , Time Factors
16.
J Trauma Acute Care Surg ; 84(4): 575-582, 2018 04.
Article in English | MEDLINE | ID: mdl-29287059

ABSTRACT

BACKGROUND: Early resuscitation after trauma-hemorrhagic shock with plasma rather than crystalloid may ameliorate systemic endothelial cell (EC) injury and dysfunction (endotheliopathy of trauma). We postulated that endothelial-lined microfluidic networks would be a useful platform to study the EC activation/injury under flow conditions to mimic trauma-hemorrhagic shock. We then used the microfluidic system to further characterize the protective effects and optimal timing of plasma infusion on the development of "endotheliopathy of trauma" in our model. METHODS: Human umbilical vein ECs were added to microfluidic flow channels, and after overnight perfusion, the cells were subsequently treated with epinephrine and exposed to hypoxia reoxygenation. Media alone or 5% human plasma was perfused either immediately following treatment (early plasma) or after a 3-hour delay (late plasma). Glycocalyx injury was indexed by fluorescent microscopy and shedding of syndecan 1 and hyaluronic acid. Endothelial markers of activation/injury were also measured and included soluble thrombomodulin, tissue plasminogen activator, plasminogen activator inhibitor 1, and angiopoietins 1 and 2. Sheddase activity was indexed by ADAM metallopeptidase domain 17. RESULTS: Endothelial cell and glycocalyx barrier function studies using microfluidic devices are a more realistic model of the glycocalyx endothelial vascular barrier than studies performed on ECs using static (no flow) conditions. Conditions that mimic the internal milieu following hemorrhagic shock result in glycocalyx degradation and an inflammatory prothrombotic response by the endothelium. "Early" use of plasma in the microfluidic channel perfusate mitigated against these effects. Later perfusion with plasma had no protective effect. CONCLUSIONS: A temporal effect to plasma administration was noted in our biomimetic model of the endothelial vascular barrier following shock. This suggests a protective role to "early" plasma administration in the severely injured patient.


Subject(s)
Endothelium, Vascular/physiopathology , Microfluidics/methods , Plasma , Resuscitation/methods , Shock, Hemorrhagic/therapy , Vasodilation/physiology , Wounds and Injuries/complications , Cells, Cultured , Human Umbilical Vein Endothelial Cells/pathology , Humans , Shock, Hemorrhagic/etiology , Shock, Hemorrhagic/physiopathology , Time Factors , Wounds and Injuries/physiopathology
18.
Am J Surg ; 214(6): 1166-1172, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28974313

ABSTRACT

BACKGROUND: Geriatric trauma patients have high circulating norepinephrine (NE) levels but attenuated release of epinephrine (Epi) in response to increasing severity of injury. We hypothesized that NE and Epi have different effects on the endothelial and glycocalyx components of the vascular barrier following shock. METHODS: Human umbilical vein endothelial cells (HUVEC) were treated with varying concentrations of NE or Epi and exposed to simulated shock conditions (HR). Relevant biomarkers were sampled to index glycocalyx injury and endothelial cell activation. RESULTS: NE was associated with significantly greater glycocalyx damage and endothelial activation/injury vs. Epi treatment groups. There were minimal changes in PAI-1 with either NE or Epi ± H/R. However NE ± H/R was associated with significantly higher tPA levels. CONCLUSIONS: NE favors a profibrinolytic state. Our study supports investigating liberal use of the anti-fibrinolytic agent tranexamic acid in the severely injured geriatric trauma patient.


Subject(s)
Epinephrine/pharmacology , Glycocalyx/metabolism , Norepinephrine/pharmacology , Shock/metabolism , Angiopoietins/metabolism , Biomarkers/metabolism , Cell Culture Techniques , Endothelial Cells , Enzyme-Linked Immunosorbent Assay , Humans , In Vitro Techniques , Plasminogen Activator Inhibitor 1/metabolism , Syndecan-1/metabolism , Thrombomodulin/metabolism , Tissue Plasminogen Activator/metabolism , Umbilical Veins/cytology
19.
J Trauma Acute Care Surg ; 82(6): 1080-1086, 2017 06.
Article in English | MEDLINE | ID: mdl-28328682

ABSTRACT

BACKGROUND: Systemic vascular endothelial injury is a consequence of trauma (T)/hemorrhagic shock (HS) which results in disturbances of coagulation, inflammation, and endothelial barrier integrity. The effect of T/HS on the endothelium (endotheliopathy of trauma [EoT]) is of intense research interest and may lead to EoT-directed therapies. Administration of tranexamic acid (TXA) in trauma patients is associated with a survival benefit and fewer complications if given early after injury. Mechanisms for this protective effect include the antifibrinolytic and anti-inflammatory effects of TXA. We hypothesized that "early" administration of TXA would abrogate vascular endothelial cell activation and injury after T/HS. This was studied in vitro. METHODS: Confluent human umbilical vein endothelial cells were exposed to hydrogen peroxide and/or epinephrine to stimulate post-T/HS oxidant exposure and/or sympathoadrenal activation. TXA was added 15 minutes, 60 minutes, or 120 minutes after H2O2 and/or epinephrine challenge. Endothelial cell injury was indexed by cell monolayer permeability, intracellular adhesion molecule expression, soluble thrombomodulin, syndecan release (marker for glycocalyx injury), tissue type plasminogen activator (tPA), plasminogen activator inhibitor-1 (PAI-1) and angiopoietin-2/angiopoietin-1 ratio (APO-2/APO-1). RESULTS: Endothelial activation and injury as indexed by permeability, ICAM expression, soluble thrombomodulin were increased by H2O2 and/or epinephrine exposure. Biomarkers of endothelial coagulation profile (tPA/PAI-1) demonstrated a profibrinolytic profile (increased tPA and tPA/PAI-1 ratio) after challenge by H2O2 and/or epinephrine. Vascular "leakiness" as indexed by APO-2/APO-1 ratio was also evident. The most profound effects were noted with H2O2/epinephrine exposure. TXA administration within 60 minutes of H2O2/epinephrine challenge abolished the adverse effects noted on the endothelial-glycocalyx "double barrier." TXA administration after 60 minutes was not protective. CONCLUSION: Antifibrinolytic and other protective effects of TXA administration on endothelial injury are time-dependent. This study supports the concept that the clinical efficacy of TXA administration requires "early administration."


Subject(s)
Antifibrinolytic Agents/therapeutic use , Endothelium, Vascular/injuries , Shock, Hemorrhagic/drug therapy , Tranexamic Acid/therapeutic use , Wounds and Injuries/drug therapy , Angiopoietin-1/metabolism , Angiopoietin-2/metabolism , Antifibrinolytic Agents/administration & dosage , Cell Adhesion Molecules/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiopathology , Epinephrine/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/physiology , Humans , Hydrogen Peroxide/pharmacology , In Vitro Techniques , Plasminogen Activator Inhibitor 1/metabolism , Shock, Hemorrhagic/physiopathology , Syndecans/metabolism , Thrombomodulin/metabolism , Tissue Plasminogen Activator/metabolism , Tranexamic Acid/administration & dosage , Wounds and Injuries/physiopathology
20.
Am J Surg ; 213(3): 489-493, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28242029

ABSTRACT

OBJECTIVE: Intraluminal pancreatic trypsin and other digestive enzymes injure the gut barrier following trauma-hemorrhagic shock (T/HS). Intestinal proteases (sheddases) exert important effects on normal gut function but may cause barrier disruption due to exaggerated production following T/HS. We hypothesized that the protective mechanism of TXA on the gut barrier following T/HS includes inhibition of these "downstream" proteases. This was studied in vitro. METHODS: Trypsin, matrix metalloproteinase (MMP-9) and ADAM-17 activity were measured in intestinal epithelial cells (IEC) exposed to HR + trypsin. TXA was added to IEC subsets. Pulmonary microvascular endothelial cells (HMVEC) were exposed to IEC supernatants and syndecan release and ICAM-1 expression determined. RESULTS: Trypsin activity and the activity of the "downstream" sheddases ADAM-17, MMP was increased in IEC lysates following exposure to HR + trypsin. Syndecan and ICAM-1 were increased in HMVEC exposed to IEC supernatants. TXA administration 'early' abrogated these effects. CONCLUSIONS: TXA administration early after shock protects the gut barrier by inhibiting trypsin uptake and activity and the subsequent downstream protease cascade. To be effective, TXA should be administered early in all "at risk" patients.


Subject(s)
Antifibrinolytic Agents/pharmacology , Tranexamic Acid/pharmacology , Trypsin/metabolism , ADAM17 Protein/drug effects , ADAM17 Protein/metabolism , Cell Line , Cells, Cultured , Epithelial Cells/metabolism , Humans , Intercellular Adhesion Molecule-1/drug effects , Intercellular Adhesion Molecule-1/metabolism , Intestines/cytology , Lung/cytology , Matrix Metalloproteinase 9/drug effects , Matrix Metalloproteinase 9/metabolism , Shock, Hemorrhagic/complications , Syndecans/metabolism , Trypsin/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...