Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(5)2024 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-38473750

RESUMEN

Uncontrolled hemorrhage is a major preventable cause of death in patients with trauma. However, the majority of large animal models of hemorrhage have utilized controlled hemorrhage rather than uncontrolled hemorrhage to investigate the impact of immunopathy and coagulopathy on multi-organ failure (MOF) and mortality. This study evaluates these alterations in a severe porcine controlled and uncontrolled hemorrhagic shock (HS) model. Anesthetized female swine underwent controlled hemorrhage and uncontrolled hemorrhage by partial splenic resection followed with or without lactated Ringer solution (LR) or Voluven® resuscitation. Swine were surveyed 6 h after completion of splenic hemorrhage or until death. Blood chemistry, physiologic variables, systemic and tissue levels of complement proteins and cytokines, coagulation parameters, organ function, and damage were recorded and assessed. HS resulted in systemic and local complement activation, cytokine release, hypocoagulopathy, metabolic acidosis, MOF, and no animal survival. Resuscitation with LR and Voluven® after HS improved hemodynamic parameters (MAP and SI), metabolic acidosis, hyperkalemia, and survival but resulted in increased complement activation and worse coagulopathy. Compared with the LR group, the animals with hemorrhagic shock treated with Voluven® had worse dilutional anemia, coagulopathy, renal and hepatic dysfunction, increased myocardial complement activation and renal damage, and decreased survival rate. Hemorrhagic shock triggers early immunopathy and coagulopathy and appears associated with MOF and death. This study indicates that immunopathy and coagulopathy are therapeutic targets that may be addressed with a high-impact adjunctive treatment to conventional resuscitation.


Asunto(s)
Acidosis , Trastornos de la Coagulación Sanguínea , Choque Hemorrágico , Humanos , Femenino , Porcinos , Animales , Insuficiencia Multiorgánica , Hemorragia , Citocinas
2.
Int J Mol Sci ; 24(8)2023 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-37108656

RESUMEN

Trauma-related hemorrhagic shock (HS) remains a leading cause of death among military and civilian trauma patients. We have previously shown that administration of complement and HMGB1 inhibitors attenuate morbidity and mortality 24 h after injury in a rat model of blast injury (BI) and HS. To further validate these results, this study aimed to develop a swine model and evaluate BI+HS-induced pathophysiology. Anesthetized Yucatan minipigs underwent combined BI and volume-controlled hemorrhage. After 30 min of shock, animals received an intravenous bolus of PlasmaLyte A and a continuous PlasmaLyte A infusion. The survival rate was 80% (4/5), and the non-survivor expired 72 min post-BI. Circulating organ-functional biomarkers, inflammatory biomarkers, histopathological evaluation, and CT scans indicated evidence of multiple-organ damage, systemic innate immunological activation, and local tissue inflammation in the injured animals. Interestingly, a rapid and dramatic increase in plasma levels of HMGB1 and C3a and markedly early myocarditis and encephalitis were associated with early death post-BI+HS. This study suggests that this model reflects the immunopathological alterations of polytrauma in humans during shock and prolonged damage control resuscitation. This experimental protocol could be helpful in the assessment of immunological damage control resuscitation approaches during the prolonged care of warfighters.


Asunto(s)
Traumatismos por Explosión , Proteína HMGB1 , Choque Hemorrágico , Humanos , Ratas , Animales , Porcinos , Modelos Animales de Enfermedad , Porcinos Enanos , Hemorragia , Choque Hemorrágico/terapia
3.
Br J Pharmacol ; 180(4): 422-440, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36251578

RESUMEN

BACKGROUND AND PURPOSE: Traumatic haemorrhage (TH) is the leading cause of potentially preventable deaths that occur during the prehospital phase of care. No effective pharmacological therapeutics are available for critical TH patients yet. Here, we identify terminal complement activation (TCA) as a therapeutic target in combat casualties and evaluate the efficacy of a TCA inhibitor (nomacopan) on organ damage and survival in vivo. EXPERIMENTAL APPROACH: Complement activation products and cytokines were analysed in plasma from 54 combat casualties. The correlations between activated complement pathway(s) and the clinical outcomes in trauma patients were assessed. Nomacopan was administered to rats subjected to lethal TH (blast injury and haemorrhagic shock). Effects of nomacopan on TH were determined using survival rate, organ damage, physiological parameters, and laboratory profiles. KEY RESULTS: Early TCA was associated with systemic inflammatory responses and clinical outcomes in this trauma cohort. Lethal TH in the untreated rats induced early TCA that correlated with the severity of tissue damage and mortality. The addition of nomacopan to a damage-control resuscitation (DCR) protocol significantly inhibited TCA, decreased local and systemic inflammatory responses, improved haemodynamics and metabolism, attenuated tissue and organ damage, and increased survival. CONCLUSION AND IMPLICATIONS: Previous findings of our and other groups revealed that early TCA represents a rational therapeutic target for trauma patients. Nomacopan as a pro-survival and organ-protective drug, could emerge as a promising adjunct to DCR that may significantly reduce the morbidity and mortality in severe TH patients while awaiting transport to critical care facilities.


Asunto(s)
Complemento C5 , Choque Hemorrágico , Ratas , Animales , Complemento C5/farmacología , Choque Hemorrágico/tratamiento farmacológico , Activación de Complemento , Factores Inmunológicos/farmacología , Fenotipo
4.
Front Immunol ; 13: 991048, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36341368

RESUMEN

Complementopathy, endotheliopathy, and coagulopathy following a traumatic injury are key pathophysiological mechanisms potentially associated with multiple-organ failure (MOF) and mortality. However, the heterogeneity in the responses of complementopathy, endotheliopathy, and coagulopathy to trauma, the nature and extent of their interplay, and their relationship to clinical outcomes remain unclear. Fifty-four poly-trauma patients were enrolled and divided into three subgroups based on their ISS. Biomarkers in blood plasma reflecting complement activation, endothelial damage, and coagulopathy were measured starting from admission to the emergency department and at 3, 6, 12, 24, and 120 hours after admission. Comparative analyses showed that severely injured patients (ISS>24) were associated with longer days on mechanical ventilation, in the intensive care unit and hospital stays, and a higher incidence of hyperglycemia, bacteremia, respiratory failure and pneumonia compared to mildly (ISS<16) or moderately (ISS=16-24) injured patients. In this trauma cohort, complement was activated early, primarily through the alternative complement pathway. As measured in blood plasma, severely injured patients had significantly higher levels of complement activation products (C3a, C5a, C5b-9, and Bb), endothelial damage markers (syndecan-1, sTM, sVEGFr1, and hcDNA), and fibrinolytic markers (D-dimer and LY30) compared to less severely injured patients. Severely injured patients also had significantly lower thrombin generation (ETP and peak) and lower levels of coagulation factors (I, V, VIII, IX, protein C) than less severely injured patients. Complement activation correlated with endothelial damage and hypocoagulopathy. Logistic regression analyses revealed that Bb >1.57 µg/ml, syndecan-1 >66.6 ng/ml or D-dimer >6 mg/L at admission were associated with a higher risk of MOF/mortality. After adjusting for ISS, each increase of the triadic score defined above (Bb>1.57 µg/ml/Syndecan-1>66.6 ng/ml/D-dimer>6.0mg/L) was associated with a 6-fold higher in the odds ratio of MOF/death [OR: 6.83 (1.04-44.96, P=0.046], and a 4-fold greater in the odds of infectious complications [OR: 4.12 (1.04-16.36), P=0.044]. These findings provide preliminary evidence of two human injury response endotypes (traumatized triad and non-traumatized triad) that align with clinical trajectory, suggesting a potential endotype defined by a high triadic score. Patients with this endotype may be considered for timely intervention to create a pro-survival/organ-protective phenotype and improve clinical outcomes.


Asunto(s)
Trastornos de la Coagulación Sanguínea , Traumatismo Múltiple , Humanos , Sindecano-1 , Puntaje de Gravedad del Traumatismo , Estudios Prospectivos , Trastornos de la Coagulación Sanguínea/etiología , Biomarcadores , Traumatismo Múltiple/complicaciones , Insuficiencia Multiorgánica
5.
Int J Mol Sci ; 23(21)2022 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-36362350

RESUMEN

Preclinical and clinical studies have shown that traumatic hemorrhage (TH) induces early complement cascade activation, leading to inflammation-associated multiple-organ dysfunction syndrome (MODS). Several previous studies have demonstrated the beneficial effects of complement inhibition in anesthetized (unconscious) animal models of hemorrhage. Anesthetic agents profoundly affect the immune response, microcirculation response, and coagulation patterns and thereby may confound the TH research data acquired. However, no studies have addressed the effect of complement inhibition on inflammation-driven MODS in a conscious model of hemorrhage. This study investigated whether early administration of decay-accelerating factor (CD55/DAF, a complement C3/C5 inhibitor) alleviates hemorrhage-induced organ damage and how DAF modulates hemorrhage-induced organ damage. DAF was administered to unanesthetized male Sprague Dawley rats subjected to pressure-controlled hemorrhage followed by a prolonged (4 h) hypotensive resuscitation with or without lactated Ringer's (LR). We assessed DAF effects on organ protection, tissue levels of complement synthesis and activation, T lymphocyte infiltration, fluid resuscitation requirements, and metabolic acidosis. Hemorrhage with (HR) or without (H) LR resuscitation resulted in significantly increased C3, C5a, and C5b-9 deposition in the lung and intestinal tissues. HR rats had significantly higher tissue levels of complement activation/deposition (particularly C5a and C5b-9 in the lung tissues), a higher but not significant amount of C3 and C5b-9 pulmonary microvascular deposition, and relatively severe injury in the lung and intestinal tissues compared to H rats. DAF treatment significantly reduced tissue C5b-9 formation and C3 deposition in the H or HR rats and decreased tissue levels of C5a and C3 mRNA in the HR rats. This treatment prevented the injury of these organs, improved metabolic acidosis, reduced fluid resuscitation requirements, and decreased T-cell infiltration in lung tissues. These findings suggest that DAF has the potential as an organ-protective adjuvant treatment for TH during prolonged damage control resuscitation.


Asunto(s)
Acidosis , Antígenos CD55 , Ratas , Masculino , Animales , Ratas Sprague-Dawley , Complejo de Ataque a Membrana del Sistema Complemento , Insuficiencia Multiorgánica/etiología , Insuficiencia Multiorgánica/prevención & control , Hemorragia , Proteínas del Sistema Complemento , Inactivadores del Complemento , Inflamación , Fenotipo
6.
Trauma Surg Acute Care Open ; 7(1): e000927, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36117727

RESUMEN

Objectives: Early complementopathy and coagulopathy are shown often after trauma. However, the prevalence of any interplay between complement cascade (ComC) and coagulation cascade (CoaC) after trauma remains unclear. This study intended to explore whether complement-coagulation crosstalk exists, which may provide a reliable guide to clinical implications in trauma patients. Methods: This single-center cohort study of trauma patients enrolled 100 patients along with 20 healthy volunteers. Blood samples from patients were collected at admission, 45, 90, 135 minutes, and 18 hours after admission. Demographic characteristics were recorded, blood levels of ComC and CoaC factors, and inflammatory cytokines were measured by ELISA, clot-based assays, or luminex multiplex assay, and partial thromboplastin (PT) and partial thromboplastin time (PTT) were assessed using a Behring blood coagulation system. Results: Compared with the healthy controls, plasma levels of complement factors (C5b-9 and Bb) and 11 tested inflammatory cytokines increased in moderately and severely injured patients as early as 45 minutes after admission and sustained higher levels up to 18 hours after admission. C5b-9 correlated positively to patients' hospital stay. In parallel, the consumption of coagulation factors I, II, X, and XIII was shown throughout the first 18 hours after admission in moderately and severely injured patients, whereas PT, PTT, D-dimer, factor VII, and factor VIII values significantly increased from the admission to 135 minutes in moderately and severely injured patients. Along with an inverse correlation between plasma Bb, factors I and II, a positive correlation between C5b-9, Bb, D-dimer, PT, and PTT was evident. Conclusions: This study demonstrates trauma-induced early activation of plasma cascades including ComC, CoaC, and fibrinolytic cascade, and their correlation between plasma cascades in severe trauma patients. Our study suggests that the simultaneous modulation of plasma cascades might benefit clinical outcomes for trauma patients. Level of evidence: Prospective study, level III.

7.
Biomolecules ; 12(1)2022 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-35053249

RESUMEN

Several preclinical and clinical reports have demonstrated that levels of circulating high mobility group box 1 protein (HMGB1) are increased early after trauma and are associated with systemic inflammation and clinical outcomes. However, the mechanisms of the interaction between HMGB1 and inflammatory mediators that lead to the development of remote organ damage after trauma remain obscure. HMGB1 and inflammatory mediators were analyzed in plasma from 54 combat casualties, collected on admission to a military hospital in Iraq, and at 8 and 24 h after admission. In total, 45 (83%) of these patients had traumatic brain injury (TBI). Nine healthy volunteers were enrolled as controls. HMGB1 plasma levels were significantly increased in the first 8 h after admission, and were found to be associated with systemic inflammatory responses, injury severity score, and presence of TBI. These data provided the rationale for designing experiments in rats subjected to blast injury and hemorrhage, to explore the effect of HMGB1 inhibition by CX-01 (2-O, 3-O desulfated heparin). Animals were cannulated, then recovered for 5-7 days before blast injury in a shock tube and volume-controlled hemorrhage. Blast injury and hemorrhage induced an early increase in HMGB1 plasma levels along with severe tissue damage and high mortality. CX-01 inhibited systemic HMGB1 activity, decreased local and systemic inflammatory responses, significantly reduced tissue and organ damage, and tended to increase survival. These data suggest that CX-01 has potential as an adjuvant treatment for traumatic hemorrhage.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Proteína HMGB1 , Animales , Lesiones Traumáticas del Encéfalo/metabolismo , Proteína HMGB1/metabolismo , Humanos , Inflamación/metabolismo , Ratas
8.
Mil Med ; 184(Suppl 1): 282-290, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30901474

RESUMEN

OBJECTIVE: Blast injury is associated with multi-organ failure (MOF), causing significant morbidity and mortality in trauma patients. However, the pathogenesis of blast-induced MOF still remains obscure. In this study, we evaluate the pathophysiological changes related to blast-induced MOF in a clinically relevant rat model of blast injury. METHODS: A moderate blast overpressure was applied to induce injury in anesthetized rats. Pathological changes were evaluated by H&E staining. Complement activation, plasminogen, and myeloperoxidase levels were analyzed by complement hemolytic assay (CH50) and/or ELISA in blood samples. RESULTS: Analysis of lung, brain, and liver tissue at 24 hour after blast overpressure revealed severe injuries. The level of complement components C3 and C1q decreased in parallel with the reduction of CH50 level in injured animals at 1, 3, and 6 hours after blast. Consumption of plasminogen was also detected as early as 1 hour post-injury. Myeloperoxidase levels were elevated within 1 hour of blast injury. CONCLUSION: Our data reveal that blast injury triggers the complement and fibrinolytic systems, which likely contribute to blast-induced MOF. Conceivably, therapies that target these systems early may improve clinical outcomes in blast patients.


Asunto(s)
Traumatismos por Explosión/tratamiento farmacológico , Fibrinolíticos/farmacocinética , Insuficiencia Multiorgánica/etiología , Animales , Traumatismos por Explosión/complicaciones , Encéfalo/efectos de los fármacos , Encéfalo/patología , Encéfalo/fisiopatología , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática/métodos , Fibrinolíticos/sangre , Fibrinolíticos/uso terapéutico , Puntaje de Gravedad del Traumatismo , Hígado/efectos de los fármacos , Hígado/patología , Hígado/fisiopatología , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/fisiopatología , Masculino , Insuficiencia Multiorgánica/tratamiento farmacológico , Ratas , Ratas Sprague-Dawley/sangre , Estadísticas no Paramétricas
9.
PLoS One ; 13(8): e0202594, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30133517

RESUMEN

BACKGROUND AND OBJECTIVE: Complement activation as an early and important inflammatory process contributes to multiple organ dysfunction after trauma. We have recently shown that complement inhibition by decay-accelerating factor (DAF) protects brain from blast-overpressure (BOP)-induced damage. This study was conducted to determine the effect of DAF on acute lung injury induced by BOP exposure and to elucidate its possible mechanisms of action. METHODS: Anesthetized adult male Sprague-Daley rats were exposed to BOP (120 kPa) from a compressed air-driven shock tube. Rats were randomly assigned to three experimental groups: 1) Control (no BOP and no DAF treatment), 2) BOP (120 kPa BOP exposure), and 3) BOP followed by treatment with rhDAF (500µg/kg, i.v) at 30 minutes after blast. After a recovery period of 3, 24, or 48 hours, animals were euthanized followed by the collection of blood and tissues at each time point. Samples were subjected to the assessment of cytokines and histopathology as well as for the interaction of high-mobility-group box 1 (HMGB1) protein, NF-κB, receptor for advanced glycation end products (RAGE), C3a, and C3aR. RESULTS: BOP exposure significantly increased in the production of systemic pro- and anti-inflammatory cytokines, and obvious pathological changes as characterized by pulmonary edema, inflammation, endothelial damage and hemorrhage in the lungs. These alterations were ameliorated by early administration of rhDAF. The rhDAF treatment not only significantly reduced the expression levels of HMGB1, RAGE, NF-κB, C3a, and C3aR, but also reversed the interaction of C3a-C3aR and nuclear translocation of HMGB1 in the lungs. CONCLUSIONS: Our findings indicate that early administration of DAF efficiently inhibits systemic and local inflammation, and mitigates blast-induced lung injury. The underlying mechanism might be attributed to its potential modulation of C3a-C3aR-HMGB1-transcriptional factor axis. Therefore, complement and/or HMGB1 may be potential therapeutic targets in amelioration of acute lung injury after blast injury.


Asunto(s)
Lesión Pulmonar Aguda/tratamiento farmacológico , Traumatismos por Explosión/tratamiento farmacológico , Antígenos CD55/administración & dosificación , Proteína HMGB1/genética , Inflamación/tratamiento farmacológico , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/fisiopatología , Animales , Traumatismos por Explosión/genética , Traumatismos por Explosión/patología , Activación de Complemento/efectos de los fármacos , Complemento C3a/antagonistas & inhibidores , Modelos Animales de Enfermedad , Humanos , Inflamación/genética , Inflamación/fisiopatología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/fisiopatología , FN-kappa B/genética , Presión/efectos adversos , Ratas , Ratas Sprague-Dawley
10.
J Surg Res ; 179(1): 153-67, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23122671

RESUMEN

BACKGROUND: Complement is invariably activated during trauma and contributes to tissue injury. Recombinant human decay-accelerating factor (DAF), a complement regulatory protein that inhibits both classical and alternative pathways, improves survival and reduces tissue damage in animal models of tissue injury. The extent to which DAF may facilitate resuscitation in hemorrhaged large animals is not known. METHODS: Male Yorkshire swine assigned to one of six groups were subjected to controlled, isobaric hemorrhage over 15 min to a target mean arterial pressure (MAP) of 35 mm Hg. Hypotension was maintained for 20 min followed by a bolus intravenous injection of DAF or vehicle followed by Hextend resuscitation. Animals were observed for 3 h after hypotensive Hextend resuscitation. Survival, blood chemistry, and physiological parameters were recorded. Additionally, tissue from lung, small intestine, liver, and kidney were subjected to histopathologic evaluation and tissue deposition of complement proteins was determined by immunohistochemistry, dot-blot, and Western blot analyses. RESULTS: Administration of DAF (25 µg/kg) to animals subjected to hemorrhage prior to Hextend infusion significantly improved survival (73% versus 27%); protected gut, lung, liver, and kidney tissue from damage; and resulted in reduced resuscitation fluid requirements when compared with animals subjected to hemorrhage and resuscitation with Hextend alone. Animals treated with a higher dose of DAF (50 µg/kg) followed by Hextend fluid resuscitation did not experience the same benefit, suggesting a narrow therapeutic range for use of DAF as adjunct to Hextend fluid. CONCLUSION: DAF improved survival and reduced early Hextend fluid resuscitation requirements in swine subjected to hemorrhagic shock. These benefits are attributed to decreased complement deposition and limited organ damage.


Asunto(s)
Antígenos CD55/farmacología , Intestino Delgado/efectos de los fármacos , Hígado/efectos de los fármacos , Pulmón/efectos de los fármacos , Resucitación , Choque Hemorrágico/complicaciones , Animales , Relación Dosis-Respuesta a Droga , Fluidoterapia , Derivados de Hidroxietil Almidón/uso terapéutico , Intestino Delgado/patología , Riñón/efectos de los fármacos , Riñón/patología , Hígado/patología , Pulmón/patología , Masculino , Modelos Animales , Resucitación/métodos , Choque Hemorrágico/mortalidad , Tasa de Supervivencia , Porcinos , Resultado del Tratamiento
11.
Shock ; 38(1): 82-91, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22683724

RESUMEN

Activation of the complement system has been associated with tissue injury after hemorrhage and resuscitation in animals. We investigated whether administration of recombinant human C1-esterase inhibitor (rhC1-INH), a regulator of complement and contact activation systems, reduces tissue damage and cytokine release and improves metabolic acidosis in a porcine model of hemorrhagic shock. Male Yorkshire swine were assigned to experimental groups and subjected to controlled, isobaric hemorrhage to a target mean arterial pressure of 35 mmHg. Hypotension was maintained for 20 min followed by a bolus intravenous injection of rhC1-INH or vehicle; animals were then observed for 3 h. Blood chemistry and physiologic parameters were recorded. Lung and small intestine tissue samples were subjected to histopathologic evaluation and immunohistochemistry to determine the extent of injury and deposition of complement proteins. Cytokine levels and quantitative assessment of renal and hepatic function were measured via enzyme-linked immunosorbent assay and chemistry analyzer, respectively. Pharmacokinetics of rhC1-INH revealed dose proportionality for maximum concentration, half-life, and the time span in which the functional C1-INH level was greater than 1 IU/mL. Recombinant human C1-INH significantly reduced renal, intestinal, and lung tissue damage in a dose-dependent manner (100 and 250 IU/kg). In addition, rhC1-INH (250 IU/kg) markedly improved hemorrhage-induced metabolic acidosis and circulating tumor necrosis factor α. The tissue-protective effects of rhC1-INH appear to be related to its ability to reduce tissue complement activation and deposition. Recombinant human C1-INH decreased tissue complement activation and deposition in hemorrhaged animals, improved metabolic acidosis, reduced circulating tumor necrosis factor α, and attenuated tissue damage in this model. The observed beneficial effects of rhC1-INH treatment on tissue injury 20 min into severe hypotension present an attractive model of low-volume resuscitation, particularly in situations with a restrictive medical logistical footprint.


Asunto(s)
Proteína Inhibidora del Complemento C1/uso terapéutico , Inactivadores del Complemento/uso terapéutico , Choque Hemorrágico/tratamiento farmacológico , Acidosis/tratamiento farmacológico , Acidosis/etiología , Animales , Presión Sanguínea/efectos de los fármacos , Activación de Complemento/efectos de los fármacos , Proteína Inhibidora del Complemento C1/administración & dosificación , Proteína Inhibidora del Complemento C1/farmacología , Inactivadores del Complemento/administración & dosificación , Inactivadores del Complemento/farmacología , Proteínas del Sistema Complemento/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Infusiones Intravenosas , Enfermedades Intestinales/etiología , Enfermedades Intestinales/prevención & control , Intestino Delgado/metabolismo , Riñón/efectos de los fármacos , Riñón/fisiopatología , Pulmón/metabolismo , Enfermedades Pulmonares/etiología , Enfermedades Pulmonares/prevención & control , Masculino , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , Daño por Reperfusión/etiología , Daño por Reperfusión/prevención & control , Choque Hemorrágico/sangre , Choque Hemorrágico/complicaciones , Choque Hemorrágico/fisiopatología , Sus scrofa , Factor de Necrosis Tumoral alfa/metabolismo
12.
J Trauma ; 71(1 Suppl): S151-60, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21795874

RESUMEN

BACKGROUND: Activation of complement system has been associated with tissue injury after hemorrhage and resuscitation in rats and swine. This study investigated whether administration of human recombinant decay-accelerating factor (DAF; a complement regulatory protein that inhibits classical and alternative pathways) reduces tissue damage in a porcine model of hemorrhagic shock. METHODS: Male Yorkshire swine assigned to four groups were subjected to controlled, isobaric hemorrhage over 15 minutes to a target mean arterial pressure of 35 mm Hg. Hypotension was maintained for 20 minutes followed by a bolus intravenous injection of DAF or vehicle and then animals were observed for 200 minutes. Blood chemistry and physiologic parameters were recorded. Tissue samples from lung and small intestine were subjected to histopathological evaluation and detection of tissue deposition of complement proteins by immunohistochemistry and Western blot analyses. RESULTS: Administration of DAF significantly reduced intestinal and lung tissue damage in a dose-dependent manner (5, 25, and 50 µg/kg). In addition, DAF treatment improved hemorrhage-induced hyperkalemia. The protective effects of DAF appear to be related to its ability to reduce tissue complement activation and deposition on affected tissues. CONCLUSIONS: DAF treatment decreased tissue complement activation and deposition in hemorrhaged animals and attenuated tissue damage at 200 minutes after treatment. The observed beneficial effects of DAF treatment on tissue injury after 20 minutes of severe hypotension presents an attractive model of small volume resuscitation, particularly in situations with a restrictive medical logistical footprint such as far-forward access to first responders in the battlefield or in remote rural or mountainous environments.


Asunto(s)
Antígenos CD55/uso terapéutico , Hiperpotasemia/prevención & control , Intestinos/patología , Pulmón/patología , Choque Hemorrágico/tratamiento farmacológico , Animales , Western Blotting , Activación de Complemento/efectos de los fármacos , Hemodinámica , Humanos , Hiperpotasemia/etiología , Intestinos/efectos de los fármacos , Pulmón/efectos de los fármacos , Masculino , Proteínas Recombinantes/uso terapéutico , Resucitación/métodos , Choque Hemorrágico/complicaciones , Choque Hemorrágico/patología , Porcinos , Factores de Tiempo
13.
J Surg Res ; 167(2): e103-15, 2011 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-20189588

RESUMEN

BACKGROUND: C-reactive protein (CRP) is an acute pro-inflammatory mediator that has been demonstrated to enhance ischemia/reperfusion (IR) injury by virtue of activating the complement system. CRP is able to interact with complement proteins such as C1q, complement factor H, and C4b-binding protein. Since complement activation is central in the expression of tissue injury following IR, we have investigated the effects of human decay-accelerating factor (DAF), a complement inhibitor, on CRP-potentiated complement activation and tissue injury in mice subjected to mesenteric IR. MATERIALS AND METHODS: Male C57B1/6 mice were allocated into eight groups: (1) Sham-operated group without IR injury; (2) CRP+Sham group; (3) IR group; (4) CRP+IR group; (5) DAF group; (6) CRP+DAF group; (7) IR+DAF group, and (8) CRP+IR+DAF group. Intestinal and lung injury, neutrophil infiltration, myeloperoxidase (MPO) expression, complement component deposition, and interleukin-6 (IL-6) production were assessed for each treatment group of mice. RESULTS: We report that administration of DAF significantly attenuates the CRP-enhanced intestinal injury as well as remote lung damages following acute mesenteric IR in mice, while DAF inhibits complement activation, suppresses neutrophil infiltration, and reduces IL-6 production. CONCLUSIONS: Our study suggests that inhibition complement activation with DAF may prove useful for the treatment of post-ischemic inflammatory injuries associated with an increased production of CRP.


Asunto(s)
Proteína C-Reactiva/metabolismo , Antígenos CD55/uso terapéutico , Mesenterio/metabolismo , Mesenterio/fisiopatología , Daño por Reperfusión/metabolismo , Daño por Reperfusión/prevención & control , Animales , Antígenos CD55/administración & dosificación , Antígenos CD55/farmacología , Complemento C5a/metabolismo , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Inyecciones Intravenosas , Interleucina-6/metabolismo , Masculino , Mesenterio/irrigación sanguínea , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Infiltración Neutrófila/efectos de los fármacos , Peroxidasa/metabolismo , Daño por Reperfusión/fisiopatología
14.
J Neuroinflammation ; 7: 24, 2010 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-20380727

RESUMEN

BACKGROUND: Activated complement system is known to mediate neuroinflammation and neurodegeneration following exposure to hypoxic-ischemic insults. Therefore, inhibition of the complement activation cascade may represent a potential therapeutic strategy for the management of ischemic brain injury. Decay-accelerating factor (DAF, also known as CD55) inhibits complement activation by suppressing the function of C3/C5 convertases, thereby limiting local generation or deposition of C3a/C5a and membrane attack complex (MAC or C5b-9) production. The present study investigates the ability of DAF to protect primary cultured neuronal cells subjected to sodium cyanide (NaCN)-induced hypoxia from degeneration and apoptosis. METHODS: Cultured primary cortical neurons from embryonic Sprague-Dawley rats were assigned one of four groups: control, DAF treatment alone, hypoxic, or hypoxic treated with DAF. Hypoxic cultures were exposed to NaCN for 1 hour, rinsed, followed by 24 hour exposure to 200 ng/ml of recombinant human DAF in normal medium. Human DAF was used in the present study and it has been shown to effectively regulate complement activation in rats. Neuronal cell function, morphology and viability were investigated by measuring plateau depolarization potential, counting the number dendritic spines, and observing TUNEL and MTT assays. Complement C3, C3a, C3a receptor (R) production, C3a-C3aR interaction and MAC formation were assessed along with the generation of activated caspase-9, activated caspase-3, and activated Src. RESULTS: When compared to controls, hypoxic cells had fewer dendritic spines, reduced plateau depolarization accompanied by increased apoptotic activity and accumulation of MAC, as well as up-regulation of C3, C3a and C3aR, enhancement of C3a-C3aR engagement, and elevated caspase and Src activity. Treatment of hypoxic cells with 200 ng/ml of recombinant human DAF resulted in attenuation of neuronal apoptosis and exerted significant protection against neuronal dendritic spine loss and plateau depolarization reduction. Furthermore, treatment with DAF resulted in decreased accumulation of C3a, MAC, C3a-C3aR interaction, caspase-9, activated caspase-3, and pTyr416-Src (activated Src) tyrosine kinase. CONCLUSION: DAF was found to reduce neuronal cell death and apoptosis in NaCN induced hypoxia. This effect is attributed to the ability of DAF to limit complement activation and inhibit the activity of Src and caspases 9 and 3. This study supports the inhibiting of complement as a neuroprotective strategy against CNS ischemia/reperfusion injury.


Asunto(s)
Antígenos CD55/farmacología , Hipoxia de la Célula/efectos de los fármacos , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Caspasa 3/metabolismo , Caspasas/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/efectos de los fármacos , Complemento C3a/metabolismo , Proteínas del Sistema Complemento/fisiología , Dendritas/fisiología , Técnica del Anticuerpo Fluorescente , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley , Receptores de Complemento/metabolismo , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/metabolismo
15.
Clin Immunol ; 130(3): 313-21, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19058762

RESUMEN

Elements of the innate and adaptive immune response have been implicated in the development of tissue damage after ischemic reperfusion (I/R). Here we demonstrate that T cells infiltrate the intestine of C57BL/6 mice subjected to intestinal I/R during the first hour of reperfusion. The intensity of the T cell infiltration was higher in B6.MRL/lpr mice subjected to intestinal I/R and reflected more severe tissue damage than that observed in control mice. Depletion of T cells limited I/R damage in B6.MRL/lpr mice, whereas repletion of B6.MRL/lpr lymph node-derived T cells into the I/R-resistant Rag-1(-/-) mouse reconstituted tissue injury. The tissue-infiltrating T cells were found to produce IL-17. Finally, IL-23 deficient mice, which are known not to produce IL-17, displayed significantly less intestinal damage when subjected to I/R. Our data assign T cells a major role in intestinal I/R damage by virtue of producing the pro-inflammatory cytokine IL-17.


Asunto(s)
Enfermedades Autoinmunes/fisiopatología , Linfocitos T CD4-Positivos/inmunología , Interleucina-17/inmunología , Intestinos , Daño por Reperfusión/fisiopatología , Animales , Enfermedades Autoinmunes/inmunología , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente , Intestinos/inmunología , Intestinos/lesiones , Intestinos/fisiopatología , Isquemia/inmunología , Isquemia/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos MRL lpr , Ratones Noqueados , Daño por Reperfusión/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA