Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
2.
J Trauma Acute Care Surg ; 95(4): 558-564, 2023 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-37314576

RESUMEN

BACKGROUND: Hemorrhage accounts for 40% of the preventable death following severe injury. Activation of systemic coagulation produces bradykinin (BK), which may cause leak from the plasma to the extravascular space and to the tissues, which is part of the complex pathophysiology of trauma-induced end-organ injury. We hypothesize that BK, released during activation of coagulation in severe injury, induces pulmonary alveolar leak. METHODS: Isolated neutrophils (PMNs) were pretreated with a specific BK receptor B2 antagonist HOE-140/icatibant and BK priming of the PMN oxidase was completed. Rats underwent tissue injury/hemorrhagic shock (TI/HS), TI/icatibant/HS, and controls (no injury). Evans blue dye was instilled, and the percentage leak from the plasma to the lung was calculated from the bronchoalveolar lavage fluid (BALF). CINC-1 and total protein were measured in the BALF, and myeloperoxidase was quantified in lung tissue. RESULTS: The BK receptor B2 antagonist HOE140/icatibant inhibited (85.0 ± 5.3%) BK priming of the PMN oxidase ( p < 0.05). The TI/HS model caused activation of coagulation by increasing plasma thrombin-antithrombin complexes ( p < 0.05). Versus controls, the TI/HS rats had significant pulmonary alveolar leak: 1.46 ± 0.21% versus 0.36 ± 0.10% ( p = 0.001) and increased total protein and CINC-1 in the BALF ( p < 0.05). Icatibant given after the TI significantly inhibited lung leak and the increase in CINC-1 in the BALF from TI/icatibant/HS rats versus TI/HS ( p < 0.002 and p < 0.05) but not the total protein. There was no PMN sequestration in the lungs. Conclusions: This mixed injury model caused systemic activation of hemostasis and pulmonary alveolar leak likely due to BK release. CONCLUSION: This mixed injury model caused systemic activation of hemostasis and pulmonary alveolar leak likely due to BK release. LEVEL OF EVIDENCE: Original Article, Basic Science.


Asunto(s)
Bradiquinina , Choque Hemorrágico , Ratas , Animales , Bradiquinina/farmacología , Bradiquinina/metabolismo , Choque Hemorrágico/complicaciones , Roedores/metabolismo , Pulmón/metabolismo , Líquido del Lavado Bronquioalveolar
3.
J Trauma Acute Care Surg ; 94(6): 857-862, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-36787438

RESUMEN

BACKGROUND: The mechanisms underlying trauma-induced coagulopathy remain elusive. Hyperfibrinolysis has been linked to increased plasminogen activation and antiprotease consumption; however, the mechanistic players in its counterpart, fibrinolysis shutdown, remain unclear. We hypothesize that thrombin-activatable fibrinolysis inhibitor (TAFI) plays a major role in fibrinolytic shutdown after injury. METHODS: As part of this observational cohort study, whole blood was collected from trauma activation patients at a single, level 1 trauma center. Citrated rapid thrombelastography and the following enzyme-linked immunosorbent assays were conducted: thrombin, antithrombin, thrombin-antithrombin complex, TAFI, plasminogen, antiplasmin, plasmin-antiplasmin (PAP), tissue plasminogen activator, plasminogen activator inhibitor 1, and tissue plasminogen activator-plasminogen activator inhibitor 1 complex. Univariate and cluster analysis were performed. RESULTS: Overall, 56 patients (median age, 33.5 years; 70% male) were included. The majority (57%) presented after blunt mechanism and with severe injury (median New Injury Severity Score, 27). Two clusters of patients were identified: Group 1 (normal fibrinolysis, n = 21) and Group 2 (fibrinolysis shutdown, n = 35). Group 2 had significantly lower fibrinolysis with a median LY30 of 1.1% (interquartile range [IQR], 0.1-1.9%) versus 2.1% (IQR, 0.5-2.8%) in Group 1; while the median LY30 was within physiologic range, 45% of patients in Group 2 were in shutdown (vs. 24% in Group 1, p = 0.09). Compared with Group 1, Group 2 had significantly higher PAP (median, 4.7 [IQR, 1.7-9.3] vs. 1.4 [1.0-2.1] µg/mL in Group 1; p = 0.002) and higher TAFI (median, 152.5% [IQR, 110.3-190.7%] vs. 121.9% [IQR, 93.2-155.6%]; p = 0.04). There was a strong correlation between PAP and TAFI ( R2 = 0.5, p = 0.0002). CONCLUSION: The presented data characterize fibrinolytic shutdown, indicating an initial plasmin burst followed by diminished fibrinolysis, which is distinct from hypofibrinolysis (inadequate plasmin burst and fibrinolysis). After an initial thrombin and plasmin burst (increased PAP), fibrinolysis is inhibited, mediated in part by increased TAFI.


Asunto(s)
Antifibrinolíticos , Trastornos de la Coagulación Sanguínea , Carboxipeptidasa B2 , Humanos , Masculino , Adulto , Femenino , Activador de Tejido Plasminógeno , Fibrinolisina , Carboxipeptidasa B2/farmacología , Inhibidor 1 de Activador Plasminogénico , Trombina , Antifibrinolíticos/farmacología , Fibrinólisis , Trastornos de la Coagulación Sanguínea/etiología , Plasminógeno
4.
J Trauma Acute Care Surg ; 94(6): 798-802, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-36805626

RESUMEN

BACKGROUND: Trauma-induced coagulopathy (TIC) has been the subject of intense study for greater than a century, and it is associated with high morbidity and mortality. The Trans-Agency Consortium for Trauma-Induced Coagulopathy, funded by the National Health Heart, Lung and Blood Institute, was tasked with developing a clinical TIC score, distinguishing between injury-induced bleeding from persistent bleeding due to TIC. We hypothesized that the Trans-Agency Consortium for Trauma-Induced Coagulopathy clinical TIC score would correlate with laboratory measures of coagulation, transfusion requirements, and mortality. METHODS: Trauma activation patients requiring a surgical procedure for hemostasis were scored in the operating room (OR) and in the first ICU day by the attending trauma surgeon. Conventional and viscoelastic (thrombelastography) coagulation assays, transfusion requirements, and mortality were correlated to the coagulation scores using the Cochran-Armitage trend test or linear regression for numerical variables. RESULTS: Increased OR TIC scores were significantly associated with abnormal conventional and viscoelastic measurements, including hyperfibrinolysis incidence, as well as with higher mortality and more frequent requirement for massive transfusion ( p < 0.0001 for all trends). Patients with OR TIC score greater than 3 were more than 31 times more likely to have an ICU TIC score greater than 3 (relative risk, 31.6; 95% confidence interval, 12.7-78.3; p < 0.0001). CONCLUSION: A clinically defined TIC score obtained in the OR reflected the requirement for massive transfusion and mortality in severely injured trauma patients and also correlated with abnormal coagulation assays. The OR TIC score should be validated in multicenter studies. LEVEL OF EVIDENCE: Prognostic and Epidemiological; Level IV.


Asunto(s)
Trastornos de la Coagulación Sanguínea , Heridas y Lesiones , Humanos , Trastornos de la Coagulación Sanguínea/diagnóstico , Trastornos de la Coagulación Sanguínea/etiología , Coagulación Sanguínea , Hemorragia/etiología , Hemostasis , Pruebas de Coagulación Sanguínea , Tromboelastografía/métodos , Heridas y Lesiones/complicaciones
5.
Shock ; 58(6): 492-497, 2022 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-36548640

RESUMEN

ABSTRACT: Background: Blood type O is the most common blood type and has lower von Willebrand factor (vWF) levels (25%-35% lower than non-O blood types). von Willebrand factor is important for initiating platelet attachment and binding factor VIII. We hypothesized that patients with type O blood are at an increased risk of trauma-induced coagulopathy and bleeding post injury. Study Design: Adult trauma activations with known blood type at a level I trauma center with field systolic blood pressure < 90 mm Hg were studied retrospectively. The relationships of blood group O versus non-O to coagulation assays, massive transfusion (MT), ventilator-free days, and mortality were adjusted for confounders. Hyperfibrinolysis (HF) was defined as thromboelastogram of percent lysis in 30 min > 3%, and fibrinolysis shutdown was defined as percent lysis in 30 min < 0.9%. von Willebrand factor activity was quantified on 212 injured patients using a STAGO apparatus. Results: Overall, 268 patients met criteria. Type O patients were more likely to develop HF than non-type O blood patients (43% vs. 29%, P = 0.06) and had significantly lower vWF activity (222% vs. 249%, P = 0.01). After adjustment for New Injury Severity Score and blunt mechanism, type O had higher odds of HF (odds ratio, 1.94, 95% confidence interval, 1.09-3.47) and increased odds of MT (odds ratio, 3.02; 95% confidence interval, 1.22-7.49). Other outcomes were not significantly affected. Conclusion: Type O patients with hypotension had increased HF and MT post injury, and these were associated with lower vWF activity. These findings have implications for the monitoring of HF in patients receiving type O whole-blood transfusions post injury.


Asunto(s)
Sistema del Grupo Sanguíneo ABO , Trastornos de la Coagulación Sanguínea , Fibrinólisis , Hemorragia , Heridas y Lesiones , Adulto , Humanos , Trastornos de la Coagulación Sanguínea/epidemiología , Transfusión Sanguínea , Hemorragia/epidemiología , Hemorragia/etiología , Puntaje de Gravedad del Traumatismo , Estudios Retrospectivos , Factores de Riesgo , Factor de von Willebrand/análisis , Heridas y Lesiones/complicaciones
6.
Transfusion ; 62(12): 2596-2608, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36309927

RESUMEN

BACKGROUND: Platelets are part of innate immunity and comprise the cellular portion of hemostasis. Platelets express sex hormone receptors on their plasma membrane and sex hormones can alter their function in vitro. Little is known about how age and sex may affect platelet biology; thus, we hypothesized that platelets from males and females have different metabolomic profiles, which may be altered by age and in vitro treatment with sex hormones. METHODS: Day 1 apheresis platelets were drawn from five 18-53-year-old, premenopausal younger females (YF), five ≥54-year-old, postmenopausal, older females (OF), five 18-44-year-old younger males (YM), and four ≥45-year-old older males (OM). Platelets were normalized to a standard concentration and metabolomics analyses were completed. Unsupervised statistical analyses and hierarchical clustering with principal component analyses were completed. RESULTS: Platelets from OM had (1) elevated mono-, di- and tri-carboxylates, (2) increased levels of free fatty acids, acyl-carnitines, and free amino acids, and (3) increased purine breakdown and deamination products. In vitro incubation with sex hormones only affected platelets from OM donors with trends towards increased ATP and other high-energy purines and decreases in L-proline and other amino acids. CONCLUSION: Platelets from OM's versus YF, OF, and YM have a different metabolome implying increased energy metabolism, more free fatty acids, acylcarnitines, and amino acids, and increased breakdown of purines and deamination products. However, only platelets from OM were affected by sex hormones in vitro. Platelets from OM are metabolically distinct, which may impart functional differences when transfused.


Asunto(s)
Ácidos Grasos no Esterificados , Metaboloma , Humanos , Persona de Mediana Edad , Proyectos Piloto , Hormonas Esteroides Gonadales , Aminoácidos
7.
Transfusion ; 62(9): 1882-1893, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35929193

RESUMEN

BACKGROUND: Female sex confers a survival advantage following severe injury in the setting of trauma-induced coagulopathy, with female platelets having heightened responsiveness likely due to estrogen. The effects of testosterone on platelet biology are unknown, and platelets express both estradiol and androgen receptors on the plasma membrane. We hypothesize testosterone decreases platelet responses in vitro, and there are baseline differences in platelet function and metabolism stratified by sex/age. STUDY DESIGN AND METHODS: Apheresis platelets were collected from: older males (OM) ≥45 years, younger males (YM) <45 years, older females (OF) ≥54 years, and younger females (YF) <54 years, and testosterone and estradiol were measured. Platelets were incubated with testosterone (5.31 ng/ml), estradiol (105 pg/ml) or vehicle and stimulated with buffer, adenosine diphosphate (20 µM), platelet activating factor (2 µM), or thrombin (0.3 U/ml). Aggregation, CD62P surface expression, fibrinogen receptor surface expression, and platelet mitochondrial metabolism were measured. RESULTS: Testosterone significantly inhibited aggregation in OF and OM (p < .05), inhibited CD41a expression in YF, YM, and OM (p < .05), and affected a few of the baseline amounts of CD62P surface expression but not platelet activation to platelet-activating factor and adenosine diphosphate, and variably changed platelet metabolism. DISCUSSION: Platelets have sex- and age-specific aggregation, receptor expression, and metabolism. Testosterone decreases platelet function dependent on the stimulus, age, and sex. Similarly, platelet metabolism has varying responses to sex hormones with baseline metabolic differences dependent upon sex and age.


Asunto(s)
Plaquetas , Agregación Plaquetaria , Adenosina Difosfato/farmacología , Plaquetas/metabolismo , Estradiol/metabolismo , Estradiol/farmacología , Femenino , Humanos , Masculino , Testosterona/farmacología
8.
Ann Surg ; 276(6): e944-e954, 2022 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33214479

RESUMEN

OBJECTIVES: Identify the metabolites that are increased in the plasma of severely injured patients that developed ARDS versus severely injured patients that did not, and assay if these increased metabolites prime pulmonary sequestration of neutrophils (PMNs) and induce pulmonary sequestration in an animal model of ARDS. We hypothesize that metabolic derangement due to advanced shock in critically injured patients leads to the PMNs, which serves as the first event in the ARDS. Summary of Background Data: Intracellular metabolites accumulate in the plasma of severely injured patients. METHODS: Untargeted metabolomics profiling of 67 critically injured patients was completed to establish a metabolic signature associated with ARDS development. Metabolites that significantly increased were assayed for PMN priming activity in vitro. The metabolites that primed PMNs were tested in a 2-event animal model of ARDS to identify a molecular link between circulating metabolites and clinical risk for ARDS. RESULTS: After controlling for confounders, 4 metabolites significantly increased: creatine, dehydroascorbate, fumarate, and succinate in trauma patients who developed ARDS ( P < 0.05). Succinate alone primed the PMN oxidase in vitro at physiologically relevant levels. Intravenous succinate-induced PMN sequestration in the lung, a first event, and followed by intravenous lipopolysaccharide, a second event, resulted in ARDS in vivo requiring PMNs. SUCNR1 inhibition abrogated PMN priming, PMN sequestration, and ARDS. Conclusion: Significant increases in plasma succinate post-injury may serve as the first event in ARDS. Targeted inhibition of the SUCNR1 may decrease ARDS development from other disease states to prevent ARDS globally.


Asunto(s)
Secuestro Broncopulmonar , Síndrome de Dificultad Respiratoria , Animales , Neutrófilos/metabolismo , Ácido Succínico/metabolismo , Secuestro Broncopulmonar/metabolismo , Pulmón
9.
J Trauma Acute Care Surg ; 92(1): 159-166, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34538821

RESUMEN

BACKGROUND: Severe injury predisposes patients to trauma-induced coagulopathy, which may be subdivided by the state of fibrinolysis. Systemic hyperfibrinolysis (HF) occurs in approximately 25% of these patients with mortality as high as 70%. Severe injury also causes the release of numerous intracellular proteins, which may affect coagulation, one of which is hemoglobin, and hemoglobin substitutes induce HF in vitro. We hypothesize that the α-globin chain of hemoglobin potentiates HF in vitro by augmenting plasmin activity. METHODS: Proteomic analysis was completed on a pilot study of 30 injured patients before blood component resuscitation, stratified by their state of fibrinolysis, plus 10 healthy controls. Different concentrations of intact hemoglobin A, the α- and ß-globin chains, or normal saline (controls) were added to whole blood, and tissue plasminogen activator (tPA)-challenged thrombelastography was used to assess the degree of fibrinolysis. Interactions with plasminogen (PLG) were evaluated using surface plasmon resonance. Tissue plasminogen activator-induced plasmin activity was evaluated in the presence of the α-globin chain. RESULTS: Only the α- and ß-globin chains increased in HF patients (p < 0.01). The α-globin chain but not hemoglobin A or the ß-globin chain decreased the reaction time and significantly increased lysis time 30 on citrated native thrombelastographies (p < 0.05). The PLG and α-globin chain had interaction kinetics similar to tPA:PLG, and the α-globin chain increased tPA-induced plasmin activity. CONCLUSIONS: The α-globin chain caused HF in vitro by binding to PLG and augmenting plasmin activity and may represent a circulating "moonlighting" mediator released by the tissue damage and hemorrhagic shock inherent to severe injury. LEVEL OF EVIDENCE: Prognostic, level III.


Asunto(s)
Trastornos de la Coagulación Sanguínea , Fibrinolisina/metabolismo , Fibrinólisis , Activador de Tejido Plasminógeno/farmacología , Heridas y Lesiones , Globinas beta/metabolismo , Adulto , Trastornos de la Coagulación Sanguínea/sangre , Trastornos de la Coagulación Sanguínea/diagnóstico , Trastornos de la Coagulación Sanguínea/etiología , Femenino , Fibrinólisis/efectos de los fármacos , Fibrinólisis/fisiología , Fibrinolíticos/farmacología , Hemoglobinas/metabolismo , Humanos , Masculino , Redes y Vías Metabólicas , Pronóstico , Proteómica/métodos , Tromboelastografía/métodos , Heridas y Lesiones/sangre , Heridas y Lesiones/complicaciones , Globinas alfa/metabolismo
10.
J Trauma Acute Care Surg ; 87(5): 1052-1060, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31162329

RESUMEN

BACKGROUND: Females are hypercoagulable and have survival benefit in trauma-induced coagulopathy (TIC). The mechanism for this sex-specific hypercoagulability is unknown. Platelets and platelet function are central in providing hemostatic potential and are the largest contributor to clot strength. Ligands (adenosine diphosphate [ADP] and platelet-activating factor [PAF]) bind distinct platelet receptors to potentiate activation and aggregation. We hypothesize that female platelets have a differential response to ADP and PAF, resulting in greater aggregation and activation compared to males, and that estradiol pretreatment of male or female platelets enhances this activity. METHODS: Platelets were collected from healthy volunteers: premenopausal/postmenopausal females (≤54 years, >54 years) and similarly aged males. Platelet aggregometry and flow cytometry (fibrinogen binding capacity) were examined. After treatment with ADP or PAF, platelet aggregation was assessed with Chronolog and activation assessed by CD41 receptor surface expression using flow cytometry. Aggregation and activation were again assessed after platelet pretreatment with estradiol. RESULTS: Healthy volunteers included 12 premenopausal and 13 postmenopausal females and 18 similarly aged males. Female platelets (combined premenopausal and postmenopausal) had increased aggregation with ADP stimulation, as compared to male platelets. Male and female platelets had differential fibrinogen receptor expression, with female platelets (combined premenopausal and postmenopausal) demonstrating robust activation with ADP versus male platelets with PAF. In the presence of estradiol incubation, male platelets' activation with PAF approximated that of females (combined premenopausal and postmenopausal) and activation with PAF was enhanced in both male and female platelets. CONCLUSION: Male and female platelets have differential response to stimuli, suggesting sex-dependent signaling and cellular activation. Female platelets have both increased aggregation and activation potential, and estradiol pretreatment feminizes male platelets to approximate female platelet activation with PAF. These findings offer potential explanation for sex-based differences in hemostatic potential in TIC and question whether donor sex of transfused platelets should be considered in resuscitation. Estradiol may also serve as a novel therapeutic adjunct in TIC.


Asunto(s)
Coagulación Sanguínea/fisiología , Plaquetas/metabolismo , Agregación Plaquetaria/fisiología , Adenosina Difosfato/metabolismo , Adulto , Anciano , Trastornos de la Coagulación Sanguínea/etiología , Trastornos de la Coagulación Sanguínea/fisiopatología , Estradiol/metabolismo , Femenino , Voluntarios Sanos , Humanos , Masculino , Persona de Mediana Edad , Factor de Activación Plaquetaria/metabolismo , Activación Plaquetaria/fisiología , Pruebas de Función Plaquetaria , Posmenopausia/sangre , Premenopausia/sangre , Receptores Fibrinógenos/metabolismo , Factores Sexuales , Heridas y Lesiones/complicaciones , Adulto Joven
11.
J Surg Res ; 225: 1-5, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29605018

RESUMEN

BACKGROUND: Plasminogen activator inhibitor-1 (PAI-1) is a major regulator of the fibrinolytic system, covalently binding to tissue plasminogen activator and blocking its activity. Fibrinolysis shutdown is evident in the majority of severely injured patients in the first 24 h and is thought to be due to PAI-1. The source of this PAI-1 is thought to be predominantly endothelial cells, but there are known organ-specific differences, with higher levels thought to be in the liver. Thrombin generation is also elevated in injured patients and is a potent stimulus for PAI-1 release in human umbilical endothelial cells. We hypothesize that thrombin induces liver endothelial cells to release increased amounts of PAI-1, versus pulmonary endothelium, consisting of both stored PAI-1 and a larger contribution from de novo PAI-1 synthesis. METHODS: Human liver sinusoidal endothelial cells (LSECs) and human microvascular lung endothelial cells (HMVECs) were stimulated in vitro ± thrombin (1 and 5 IU/mL) for 15-240 min, the supernatants were collected, and PAI-1 was measured by enzyme-linked immunosorbent assays. To elucidate the PAI-1 contribution from storage versus de novo synthesis, cycloheximide (10 µg/mL) was added before thrombin in separate experiments. RESULTS: While both LSECs and HMVECs rapidly stimulated PAI-1 release, LSECs released more PAI-1 than HMVECs in response to high-dose thrombin, whereas low-dose thrombin did not provoke immediate release. LSECs continued to release PAI-1 over the ensuing 240 min, whereas HMVECs did not. Cycloheximide did not inhibit early PAI-1 release from LSECs but did at the later time points (30-240 min). CONCLUSIONS: Thrombin elicits increased amounts of PAI-1 release from liver endothelium compared with lung, with a small presynthesized stored contribution and a later, larger increase in PAI-1 release via de novo synthesis. This study suggests that the liver may be an important therapeutic target for inhibition of the hypercoagulable surgical patient and the associated complications that result.


Asunto(s)
Células Endoteliales/metabolismo , Fibrinólisis/fisiología , Hígado/metabolismo , Pulmón/metabolismo , Inhibidor 1 de Activador Plasminogénico/metabolismo , Trombina/metabolismo , Células Cultivadas , Cicloheximida/farmacología , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Ensayo de Inmunoadsorción Enzimática , Humanos , Hígado/citología , Pulmón/citología , Inhibidor 1 de Activador Plasminogénico/análisis , Biosíntesis de Proteínas/efectos de los fármacos , Inhibidores de la Síntesis de la Proteína/farmacología , Trombosis/etiología , Trombosis/fisiopatología , Activador de Tejido Plasminógeno/metabolismo , Heridas y Lesiones/complicaciones , Heridas y Lesiones/fisiopatología
12.
Transfusion ; 58(4): 891-895, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29473178

RESUMEN

BACKGROUND: Platelets (PLTs) collected and stored in PLT additive solution Intersol (PAS-C) are presumed to reduce recipient exposure to donor plasma components; however, the effects of PAS-C on PLT supernatant composition are poorly defined. Therefore, we compared the total protein concentration, isohemagglutinin titers, and HLA antibodies in supernatants of PAS-C PLTs to plasma PLTs. STUDY DESIGN AND METHODS: Apheresis PLTs from group O blood donors were collected into either 100% donor plasma (n = 50) or a mixture of 65% PAS-C/35% donor plasma (n = 50). Within 12 hours of collection, samples of the PLT supernatant were frozen and stored. PLT supernatants were assayed for total protein concentration and anti-A and anti-B titers and screened for HLA antibodies. Samples positive for HLA antibodies were further tested using single-antigen bead assays to determine antibody strength and specificity. RESULTS: Supernatants of PAS-C PLTs had significantly lower total protein concentration and anti-A and anti-B titers compared to plasma PLTs. There was no significant difference in the number of HLA antibody screen-positive PAS-C (3/50) compared to plasma PLT supernatants (2/50); however, the HLA antibody screen-positive supernatants of PAS-C PLTs had fewer HLA specificities (2) compared to those of the plasma PLTs (18). CONCLUSION: Decreased plasma proteins likely underlie lower rates of allergic and febrile, nonhemolytic transfusion reactions from the infusion of PAS-C PLTs. Decreased anti-A and anti-B titers may prevent hemolysis from minor ABO mismatch. Lower HLA antibody specificities may mitigate transfusion-related acute lung injury.


Asunto(s)
Plaquetas/efectos de los fármacos , Conservación de la Sangre/métodos , Proteínas Sanguíneas/análisis , Hemaglutininas/sangre , Prueba de Histocompatibilidad/métodos , Isoanticuerpos/sangre , Soluciones Preservantes de Órganos/farmacología , Lesión Pulmonar Aguda Postransfusional/prevención & control , Sistema del Grupo Sanguíneo ABO/inmunología , Afinidad de Anticuerpos , Especificidad de Anticuerpos , Incompatibilidad de Grupos Sanguíneos/prevención & control , Plaquetas/química , Plaquetas/inmunología , Epítopos/inmunología , Femenino , Antígenos HLA/inmunología , Humanos , Masculino , Plasma , Plaquetoferesis
13.
Transfusion ; 57(11): 2690-2700, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28880373

RESUMEN

BACKGROUND: Although transfusion is a lifesaving intervention, it may be associated with significant morbidity in injured patients. We hypothesize that stored red blood cells (RBCs) induce proinflammatory activation of human pulmonary microvascular endothelial cells (HMVECs) resulting in neutrophil (PMN) adhesion and predisposition to acute lung injury (ALI). STUDY DESIGN AND METHODS: Ten units of RBCs were collected; 50% (by weight) were leukoreduced (LR-RBCs) and the remainder was unmodified and stored in additive solution-5 (AS-5). An additional 10 units of RBCs were collected, leukoreduced, and stored in AS-3. HMVECs were incubated with [10%-40%]FINAL of the supernatants on Day (D)1 to D42 of storage, lipid extracts, and purified lipids. Endothelial surface expression of intercellular adhesion molecule-1 (ICAM-1), interleukin (IL)-8 release, and PMN adhesion to HMVECs were measured. HMVEC signaling via the BLT2 receptor was evaluated. Supernatants and lipids were also employed as the first event in a two-event model of ALI. RESULTS: The supernatants [10%-40%]FINAL from D21 LR-RBCs and D42 RBCs and LR-RBCs and the lipids from D42 stored in AS-5 induced increased ICAM-1 surface expression on endothelium, IL-8 release, and PMN adhesion. In addition, the supernatants [20%-40%]FINAL from D21 and D42 RBCs in AS-5 also increased endothelial surface expression of ICAM-1. D42 supernatants and lipids also caused coprecipitation of ß-arrestin-1 with BLT2, protein kinase C (PKC)ßI , and PKCδ and served as the first event in a two-event rodent model of ALI. CONCLUSION: Lipids that accumulate during RBC storage activate endothelium and predispose to ALI, which may explain some of the adverse events associated with the transfusion of critically injured patients.


Asunto(s)
Conservación de la Sangre/métodos , Eritrocitos/citología , Lípidos/farmacología , Pulmón/irrigación sanguínea , Proteína Quinasa C/metabolismo , Receptores de Leucotrieno B4/metabolismo , Lesión Pulmonar Aguda/etiología , Medios de Cultivo Condicionados/farmacología , Células Endoteliales/metabolismo , Activación Enzimática , Transfusión de Eritrocitos/efectos adversos , Humanos , Microvasos/citología , Neumonía/etiología
14.
Blood Transfus ; 15(2): 131-136, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28263170

RESUMEN

Pre-storage leucoreduction has been universally adopted in most developed countries in Asia, Europe and the Americas. It decreases febrile transfusion reactions, alloimmunisation to HLA antigens, cytomegalovirus exposure, the accumulation of a number of pro-inflammatory mediators in the supernatant, including the accumulation of platelet-and leucocyte-derived proteins and metabolites during routine storage. This review will highlight the lipids and proteins, biological response modifiers (BRMs) that accumulate, their clinical effects in transfused hosts, and methods of mitigation.


Asunto(s)
Lesión Pulmonar Aguda , Conservación de la Sangre/efectos adversos , Proteínas Sanguíneas/metabolismo , Transfusión de Eritrocitos/efectos adversos , Eritrocitos/metabolismo , Procedimientos de Reducción del Leucocitos , Metabolismo de los Lípidos , Lesión Pulmonar Aguda/sangre , Lesión Pulmonar Aguda/etiología , Lesión Pulmonar Aguda/terapia , Conservación de la Sangre/métodos , Transfusión de Eritrocitos/métodos , Humanos
15.
J Leukoc Biol ; 101(1): 261-273, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27531930

RESUMEN

Lysophosphatidylcholines (lysoPCs) are effective polymorphonuclear neutrophil (PMN) priming agents implicated in transfusion-related acute lung injury (TRALI). LysoPCs cause ligation of the G2A receptor, cytosolic Ca2+ flux, and activation of Hck. We hypothesize that lysoPCs induce Hck-dependent activation of protein kinase C (PKC), resulting in phosphorylation and membrane translocation of 47 kDa phagocyte oxidase protein (p47phox). PMNs, human or murine, were primed with lysoPCs and were smeared onto slides and examined by digital microscopy or separated into subcellular fractions or whole-cell lysates. Proteins were immunoprecipitated or separated by polyacrylamide gel electrophoresis and immunoblotted for proteins of interest. Wild-type (WT) and PKCγ knockout (KO) mice were used in a 2-event model of TRALI. LysoPCs induced Hck coprecipitation with PKCδ and PKCγ and the PKCδ:PKCγ complex also had a fluorescence resonance energy transfer (FRET)+ interaction with lipid rafts and Wiskott-Aldrich syndrome protein family verprolin-homologous protein 2 (WAVE2). PKCγ then coprecipitated with p47phox Immunoblotting, immunoprecipitation (IP), specific inhibitors, intracellular depletion of PKC isoforms, and PMNs from PKCγ KO mice demonstrated that Hck elicited activation/Tyr phosphorylation (Tyr311 and Tyr525) of PKCδ, which became Thr phosphorylated (Thr507). Activated PKCδ then caused activation of PKCγ, both by Tyr phosphorylation (Τyr514) and Ser phosphorylation, which induced phosphorylation and membrane translocation of p47phox In PKCγ KO PMNs, lysoPCs induced Hck translocation but did not evidence a FRET+ interaction between PKCδ and PKCγ nor prime PMNs. In WT mice, lysoPCs served as the second event in a 2-event in vivo model of TRALI but did not induce TRALI in PKCγ KO mice. We conclude that lysoPCs prime PMNs through Hck-dependent activation of PKCδ, which stimulates PKCγ, resulting in translocation of phosphorylated p47phox.


Asunto(s)
Membrana Celular/metabolismo , Lisofosfatidilcolinas/farmacología , NADPH Oxidasas/metabolismo , Neutrófilos/metabolismo , Proteína Quinasa C-delta/metabolismo , Proteína Quinasa C/metabolismo , Proteínas Proto-Oncogénicas c-hck/metabolismo , Animales , Calcio/metabolismo , Membrana Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Humanos , Lesión Pulmonar/patología , Ratones , Ratones Noqueados , Neutrófilos/efectos de los fármacos , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Transporte de Proteínas/efectos de los fármacos , Proteínas Recombinantes/farmacología
16.
Transfusion ; 56(12): 3004-3011, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27667662

RESUMEN

BACKGROUND: Transfusion-related acute lung injury (TRALI) is a significant cause of mortality, especially after transfusions containing antibodies to major histocompatibility complex (MHC) class II antigens. We hypothesize that a first event induces both 1) polymorphonuclear neutrophils (PMNs) to express MHC class II antigens, and 2) activation of the pulmonary endothelium, leading to PMN sequestration, so that the infusion of specific MHC class II antibodies to these antigens causes PMN-mediated acute lung injury (ALI). STUDY DESIGN AND METHODS: Rats were treated with saline (NS), endotoxin (lipopolysaccharide [LPS]), or cytokines (interferon-γ [IFNγ], macrophage colony-stimulating factor [MCSF], tumor necrosis factor-α [TNFα]); the PMNs were isolated; and the surface expression of the MHC class II antigen OX6 and priming by OX6 antibodies were measured by flow cytometry or priming assays. RESULTS: A two-event model of ALI was completed with NS, LPS, or IFNγ/MCSF/TNFα (first events) and the infusion of OX6 (second event). Compared with NS incubation, rats treated with either LPS or IFNγ/MCSF/TNFα exhibited OX6 PMN surface expression, OX6 antibodies primed the formyl-methionyl-leucyl phenylalanine (fMLF)-activated respiratory burst, and PMN sequestration was increased. OX6 antibody infusion into LPS-incubated or IFNγ/MCSF/TNFα-incubated rats elicited ALI, the OX6 antibody was present on the PMNs, and PMN depletion abrogated ALI. CONCLUSION: Proinflammatory first events induce PMN MHC class II surface expression, activation of the pulmonary endothelium, and PMN sequestration such that the infusion of cognate antibodies precipitates TRALI.


Asunto(s)
Lesión Pulmonar Aguda/etiología , Anticuerpos/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Activación Neutrófila/inmunología , Neutrófilos/inmunología , Lesión Pulmonar Aguda/patología , Animales , Citocinas/farmacología , Endotelio/patología , Antígenos de Histocompatibilidad Clase II/genética , Lipopolisacáridos/farmacología , Activación Neutrófila/efectos de los fármacos , Neutrófilos/metabolismo , Ratas , Estallido Respiratorio/inmunología , Reacción a la Transfusión
17.
Pediatr Transplant ; 20(5): 658-66, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27114335

RESUMEN

HSCT is a lifesaving procedure for children with malignant and non-malignant conditions. The conditioning regimen renders the patient severely immunocompromised and recovery starts with neutrophil (PMN) engraftment. We hypothesize that children demonstrate minimal PMN dysfunction at engraftment and beyond, which is influenced by the stem cell source and the conditioning regimen. Peripheral blood was serially collected from children at 1 to 12 months following allogeneic HSCT. PMN superoxide (O2-) production, degranulation (elastase), CD11b surface expression, and phagocytosis were assessed. Twenty-five patients, mean age of 10.5 yr with 65% males, comprised the study and transplant types included: 14 unrelated cord blood stem cells (cords), seven matched related bone marrow donors, three matched unrelated bone marrow donors, and one peripheral blood progenitor cells. Engraftment occurred at 24 days. There were no significant differences between controls and patients in PMN O2- production, phagocytosis, CD11b surface expression, and total PMN elastase. Elastase release was significantly decreased <6 months vs. controls (p < 0.05) and showed normalization by six months for cords only. The conditioning regimen did not affect PMN function. PMN function returns with engraftment, save elastase release, which occurs later related to the graft source utilized, and its clinical significance is unknown.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Neutrófilos/fisiología , Adolescente , Biomarcadores/sangre , Estudios de Casos y Controles , Niño , Preescolar , Femenino , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Lactante , Masculino , Evaluación de Resultado en la Atención de Salud , Elastasa Pancreática/sangre , Fagocitosis , Periodo Posoperatorio , Factores de Tiempo , Acondicionamiento Pretrasplante/métodos , Adulto Joven
18.
Shock ; 44(2): 137-42, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25944790

RESUMEN

UNLABELLED: Proinflammatory activation of vascular endothelium leading to increased surface expression of adhesion molecules and neutrophil (PMN) sequestration and subsequent activation is paramount in the development of acute lung injury and organ injury in injured patients. We hypothesize that α-enolase, which accumulates in injured patients, primes PMNs and causes proinflammatory activation of endothelial cells leading to PMN-mediated cytotoxicity. METHODS: Proteomic analyses of field plasma samples from injured versus healthy patients were used for protein identification. Human pulmonary microvascular endothelial cells (HMVECs) were incubated with α-enolase or thrombin, and intercellular adhesion molecule-1 surface expression was measured by flow cytometry. A two-event in vitro model of PMN cytotoxicity HMVECs activated with α-enolase, thrombin, or buffer was used as targets for lysophosphatidylcholine-primed or buffer-treated PMNs. The PMN priming activity of α-enolase was completed, and lysates from both PMNs and HMVECs were immunoblotted for protease-activated receptor 1 (PAR-1) and PAR-2 and coprecipitation of α-enolase with PAR-2 and plasminogen/plasmin. RESULTS: α-Enolase increased 10.8-fold in injured patients (P < 0.05). Thrombin and α-enolase significantly increased intercellular adhesion molecule-1 surface expression on HMVECs, which was inhibited by antiproteases, induced PMN adherence, and served as the first event in the two-event model of PMN cytotoxicity. α-Enolase coprecipitated with PAR-2 and plasminogen/plasmin on HMVECs and PMNs and induced PMN priming, which was inhibited by tranexamic acid, and enzymatic activity was not required. CONCLUSIONS: α-Enolase increases after injury and may activate pulmonary endothelial cells and prime PMNs through plasmin activity and PAR-2 activation. Such proinflammatory endothelial activation may predispose to PMN-mediated organ injury.


Asunto(s)
Células Endoteliales/metabolismo , Pulmón/irrigación sanguínea , Microcirculación , Neutrófilos/metabolismo , Fosfopiruvato Hidratasa/metabolismo , Plasminógeno/metabolismo , Receptor PAR-2/metabolismo , Lesión Pulmonar Aguda/metabolismo , Membrana Celular/metabolismo , Células Endoteliales/citología , Citometría de Flujo , Humanos , Inflamación , Molécula 1 de Adhesión Intercelular/metabolismo , Proteómica , Trombina/metabolismo , Ácido Tranexámico/metabolismo , Heridas no Penetrantes/sangre
19.
J Proteomics ; 112: 190-209, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25201077

RESUMEN

Proteomics has identified potential pathways involved in platelet storage lesions, which correlate with untoward effects in the recipient, including febrile non-haemolytic reactions. We hypothesize that an additional pathway involves protein mediators that accumulate in the platelet supernatants during routine storage in a donor gender-specific fashion. Apheresis platelet concentrates were collected from 5 healthy males and 5 females and routinely stored. The 14 most abundant plasma proteins were removed and the supernatant proteins from days 1 and 5 were analyzed via 1D-SDS-PAGE/nanoLC-MS/MS, before label-free quantitative proteomics analyses. Findings from a subset of 18 proteins were validated via LC-SRM analyses against stable isotope labeled standards. A total of 503 distinct proteins were detected in the platelet supernatants from the 4 sample groups: female or male donor platelets, either at storage day 1 or 5. Proteomics suggested a storage and gender-dependent impairment of blood coagulation mediators, pro-inflammatory complement components and cytokines, energy and redox metabolic enzymes. The supernatants from female donors demonstrated increased deregulation of structural proteins, extracellular matrix proteins and focal adhesion proteins, possibly indicating storage-dependent platelet activation. Routine storage of platelet concentrates induces changes in the supernatant proteome, which may have effects on the transfused patient, some of which are related to donor gender. BIOLOGICAL SIGNIFICANCE: The rationale behind this study is that protein components in platelet releasates have been increasingly observed to play a key role in adverse events and impaired homeostasis in transfused recipients. In this view, proteomics has recently emerged as a functional tool to address the issue of protein composition of platelet releasates from buffy coat-derived platelet concentrates in the blood bank. Despite early encouraging studies on buffy coat-derived platelet concentrates, platelet releasates from apheresis platelets have not been hitherto addressed by means of extensive proteomics technologies. Indeed, apheresis platelets are resuspended in donors' plasma, which hampers detection of less abundant proteins, owing to the overwhelming abundance of albumin (and a handful of other proteins), and the dynamic range of protein concentrations of plasma proteins. In order to cope with these issues, we hereby performed an immuno-affinity column-based depletion of the 14 most abundant plasma proteins. Samples were thus assayed via GeLC-MS, a workflow that allowed us to cover an unprecedented portion of the platelet supernatant proteome, in comparison to previous transfusion medicine-oriented studies in the literature. Finally, we hereby address the issue of biological variability, by considering the donor gender as a key factor influencing the composition of apheresis platelet supernatants. As a result, we could conclude that platelet supernatants from male and female donors are not only different in the first place, but they also store differently. This conclusion has been so far only suggested by classic transfusion medicine studies, but has been hitherto unsupported by actual biochemistry/proteomics investigations. In our opinion, the main strengths of this study are related to the analytical workflow (immunodepletion and GeLC-MS) and proteome coverage, the translational validity of the results (from a transfusion medicine standpoint) and the biological conclusion about the intrinsic (and storage-dependent) gender-related differences of platelet supernatants.


Asunto(s)
Plaquetas/metabolismo , Conservación de la Sangre , Proteínas Sanguíneas/metabolismo , Plaquetoferesis , Proteómica , Caracteres Sexuales , Plaquetas/citología , Femenino , Humanos , Masculino , Persona de Mediana Edad
20.
Blood ; 123(22): 3488-95, 2014 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-24747436

RESUMEN

Transfusion-related acute lung injury (TRALI) remains a significant cause of transfusion-related mortality with red cell transfusion. We hypothesize that prestorage filtration may reduce proinflammatory activity in the red blood cell (RBC) supernatant and prevent TRALI. Filters were manufactured for both small volumes and RBC units. Plasma containing antibodies to human lymphocyte antigen (HLA)-A2 or human neutrophil antigen (HNA)-3a was filtered, and immunoglobulins and specific HNA-3a and HLA-2a neutrophil (PMN) priming activity were measured. Antibodies to OX27 were added to plasma, and filtration was evaluated in a 2-event animal model of TRALI. RBC units from 31 donors known to have antibodies against HLA antigens and from 16 antibody-negative controls were filtered. Furthermore, 4 RBC units were drawn and underwent standard leukoreduction. Immunoglobulins, HLA antibodies, PMN priming activity, and the ability to induce TRALI in an animal model were measured. Small-volume filtration of plasma removed >96% of IgG, antibodies to HLA-A2 and HNA-3a, and their respective priming activity, as well as mitigating antibody-mediated in vivo TRALI. In RBC units, experimental filtration removed antibodies to HLA antigens and inhibited the accumulation of lipid priming activity and lipid-mediated TRALI. We conclude that filtration removes proinflammatory activity and the ability to induce TRALI from RBCs and may represent a TRALI mitigation step.


Asunto(s)
Lesión Pulmonar Aguda/etiología , Anticuerpos/inmunología , Eliminación de Componentes Sanguíneos/métodos , Transfusión de Componentes Sanguíneos/efectos adversos , Eritrocitos/inmunología , Filtración/métodos , Lípidos/inmunología , 2,3-Difosfoglicerato/sangre , Adenosina Trifosfato/sangre , Animales , Donantes de Sangre , Femenino , Antígenos HLA/inmunología , Humanos , Concentración de Iones de Hidrógeno , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Masculino , Plasma/inmunología , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...