Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Semin Immunol ; 28(3): 278-84, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27157701

RESUMEN

After severe trauma, the immune system is challenged with a multitude of endogenous and exogenous danger molecules. The recognition of released danger patterns is one of the prime tasks of the innate immune system. In the last two decades, numerous studies have established the complement cascade as a major effector system that detects and processes such danger signals. Animal models with engineered deficiencies in certain complement proteins have demonstrated that widespread complement activation after severe injury culminates in complement dysregulation and excessive generation of complement activation fragments. Such exuberant pro-inflammatory signaling evokes systemic inflammation, causes increased susceptibility to infections and is associated with a detrimental course of the disease after injury. We discuss the underlying processes of such complementopathy and recapitulate different intervention strategies within the complement cascade. So far, several orthogonal anti-complement approaches have been tested with varying success in a large number of rodent, in several porcine and few simian studies. We illustrate the different features among those intervention strategies and highlight those that hold the greatest promise to become potential therapeutic options for the intricate disease of traumatic injury.


Asunto(s)
Inactivadores del Complemento/uso terapéutico , Inmunoterapia/métodos , Inflamación/terapia , Choque Hemorrágico/terapia , Heridas y Lesiones/terapia , Animales , Modelos Animales de Enfermedad , Humanos , Inmunidad Innata , Inflamación/inmunología , Ratones , Ratones Noqueados , Moléculas de Patrón Molecular Asociado a Patógenos/inmunología , Choque Hemorrágico/inmunología , Heridas y Lesiones/inmunología
2.
J Immunol ; 198(12): 4846-4854, 2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28490576

RESUMEN

During sepsis, excessive activation of the complement system with generation of the anaphylatoxin C5a results in profound disturbances in crucial neutrophil functions. Moreover, because neutrophil activity is highly dependent on intracellular pH (pHi), we propose a direct mechanistic link between complement activation and neutrophil pHi In this article, we demonstrate that in vitro exposure of human neutrophils to C5a significantly increased pHi by selective activation of the sodium/hydrogen exchanger. Upstream signaling of C5a-mediated intracellular alkalinization was dependent on C5aR1, intracellular calcium, protein kinase C, and calmodulin, and downstream signaling regulated the release of antibacterial myeloperoxidase and lactoferrin. Notably, the pH shift caused by C5a increased the glucose uptake and activated glycolytic flux in neutrophils, resulting in a significant release of lactate. Furthermore, C5a induced acidification of the extracellular micromilieu. In experimental murine sepsis, pHi of blood neutrophils was analogously alkalinized, which could be normalized by C5aR1 inhibition. In the clinical setting of sepsis, neutrophils from patients with septic shock likewise exhibited a significantly increased pHi These data suggest a novel role for the anaphylatoxin C5a as a master switch of the delicate pHi balance in neutrophils resulting in profound inflammatory and metabolic changes that contribute to hyperlactatemia during sepsis.


Asunto(s)
Activación de Complemento , Complemento C5a/metabolismo , Activación Neutrófila , Neutrófilos/inmunología , Sepsis/inmunología , Sepsis/metabolismo , Animales , Antiácidos/farmacología , Calcio/metabolismo , Calmodulina/metabolismo , Complemento C5a/inmunología , Glucosa/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Lactatos/metabolismo , Lactoferrina , Ratones , Neutrófilos/química , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Peroxidasa/metabolismo , Proteína Quinasa C/inmunología , Proteína Quinasa C/metabolismo , Receptor de Anafilatoxina C5a/metabolismo , Transducción de Señal
3.
Arch Toxicol ; 92(1): 323-336, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28924833

RESUMEN

Bacterial protein toxins became valuable molecular tools for the targeted modulation of cell functions in experimental pharmacology and attractive therapeutics because of their potent and specific mode of action in human cells. C2IN-C3lim, a recombinant fusion toxin (~50 kDa) of the Rho-inhibiting C3lim from Clostridium (C.) limosum and a non-toxic portion of the C. botulinum C2 toxin (C2IN), is selectively internalized into the cytosol of monocytic cells where C3lim specifically ADP-ribosylates Rho A and -B, thereby inhibiting Rho-mediated signaling. Thus, we hypothesized that these unique features make C2IN-C3lim an attractive molecule for the targeted pharmacological down-regulation of Rho-mediated functions in monocytes. The analysis of the actin structure and the Rho ADP-ribosylation status implied that C2IN-C3lim entered the cytosol of primary human monocytes from healthy donors ex vivo within 1 h. Moreover, it inhibited the fMLP-induced chemotaxis of human monocytes in a Boyden chamber model ex vivo. Similarly, in a 3-dimensional ex vivo model of extravasation, single cell analysis revealed that C2IN-C3lim-treated cells were not able to move. In a clinically relevant mouse model of blunt chest trauma, the local application of C2IN-C3lim into the lungs after thorax trauma prevented the trauma-induced recruitment of monocytes into the lungs in vivo. Thus, C2IN-C3lim might be an attractive lead compound for novel pharmacological strategies to avoid the cellular damage response caused by monocytes in damaged tissue after trauma and during systemic inflammation. The results suggest that the pathophysiological role of clostridial C3 toxins might be a down-modulation of the innate immune system.


Asunto(s)
ADP Ribosa Transferasas/genética , Toxinas Botulínicas/genética , Quimiotaxis/efectos de los fármacos , Monocitos/efectos de los fármacos , Proteínas Recombinantes de Fusión/farmacología , Proteínas de Unión al GTP rho/antagonistas & inhibidores , Animales , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Pulmón/efectos de los fármacos , Pulmón/patología , Macrófagos/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Monocitos/citología , Proteínas Recombinantes de Fusión/genética , Traumatismos Torácicos/tratamiento farmacológico , Heridas no Penetrantes/tratamiento farmacológico , Proteínas de Unión al GTP rho/metabolismo
4.
Mediators Inflamm ; 2018: 2052356, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30002598

RESUMEN

BACKGROUND: Polymorphonuclear granulocytes (PMN) play a crucial role in host defense. Physiologically, exposure of PMN to the complement activation product C5a results in a protective response against pathogens, whereas in the case of systemic inflammation, excessive C5a substantially impairs neutrophil functions. To further elucidate the inability of PMN to properly respond to C5a, this study investigates the role of the cellular membrane potential of PMN in response to C5a. METHODS: Electrophysiological changes in cellular and mitochondrial membrane potential and intracellular pH of PMN from human healthy volunteers were determined by flow cytometry after exposure to C5a. Furthermore, PMN from male Bretoncelles-Meishan-Willebrand cross-bred pigs before and three hours after severe hemorrhagic shock were analyzed for their electrophysiological response. RESULTS: PMN showed a significant dose- and time-dependent depolarization in response to C5a with a strong response after one minute. The chemotactic peptide fMLP also evoked a significant shift in the membrane potential of PMN. Acidification of the cellular microenvironment significantly enhanced depolarization of PMN. In a clinically relevant model of porcine hemorrhagic shock, the C5a-induced changes in membrane potential of PMN were markedly diminished compared to healthy littermates. Overall, these membrane potential changes may contribute to PMN dysfunction in an inflammatory environment.


Asunto(s)
Complemento C5a/farmacología , Potenciales de la Membrana/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Choque Hemorrágico/metabolismo , Animales , Células Cultivadas , Relación Dosis-Respuesta a Droga , Electrofisiología , Citometría de Flujo , Humanos , Concentración de Iones de Hidrógeno , Masculino , Porcinos
5.
J Immunol ; 192(11): 5324-31, 2014 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-24790148

RESUMEN

Sepsis and septic shock, caused by an excessive systemic host-inflammatory response, are associated with high morbidity and mortality. The complement system and TLRs provide important pattern recognition receptors initiating the cytokine storm by extensive cross-talk. We hypothesized that double blockade of complement C5 and the TLR coreceptor CD14 could improve survival of experimental polymicrobial sepsis. Mice undergoing cecal ligation and puncture (CLP)-induced sepsis were treated with neutralizing anti-CD14 Ab biG 53, complement C5 inhibitor coversin (Ornithodoros moubata C inhibitor), or a combination thereof. The inflammatory study (24-h observation) revealed statistically significant increases in 22 of 24 measured plasma biomarkers in the untreated CLP group, comprising 14 pro- and anti-inflammatory cytokines and 8 chemokines, growth factors, and granulocyte activation markers. Single CD14 or C5 blockade significantly inhibited 20 and 19 of the 22 biomarkers, respectively. Combined CD14 and C5 inhibition significantly reduced all 22 biomarkers (mean reduction 85%; range 54-95%) compared with the untreated CLP group. Double blockade was more potent than single treatment and was required to significantly inhibit IL-6 and CXCL1. Combined inhibition significantly reduced morbidity (motility and eyelid movement) and mortality measured over 10 d. In the positive control CLP group, median survival was 36 h (range 24-48 h). Combined treatment increased median survival to 96 h (range 24-240 h) (p = 0.001), whereas survival in the single-treatment groups was not significantly increased (median and range for anti-CD14 and anti-C5 treatment were 36 h [24-48 h] and 48 h [24-96 h]). Combined with standard intervention therapy, specific blockade of CD14 and C5 might represent a promising new therapeutic strategy for treatment of polymicrobial sepsis.


Asunto(s)
Anticuerpos Neutralizantes/farmacología , Complemento C5/antagonistas & inhibidores , Complemento C5/inmunología , Citocinas/inmunología , Receptores de Lipopolisacáridos/inmunología , Sepsis/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Masculino , Ratones , Sepsis/tratamiento farmacológico , Sepsis/microbiología , Factores de Tiempo
6.
J Immunol ; 190(8): 4215-25, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23479227

RESUMEN

During experimental sepsis, excessive generation of the anaphylatoxin C5a results in reduction of the C5a receptor (C5aR) on neutrophils. These events have been shown to result in impaired innate immunity. However, the regulation and fate of C5aR on neutrophils during sepsis are largely unknown. In contrast to 30 healthy volunteers, 60 patients in septic shock presented evidence of complement activation with significantly increased serum levels of C3a, C5a, and C5b-9. In the septic shock group, the corresponding decrease in complement hemolytic activity distinguished survivors from nonsurvivors. Neutrophils from patients in septic shock exhibited decreased C5aR expression, which inversely correlated with serum concentrations of C-reactive protein (CRP) and clinical outcome. In vitro exposure of normal neutrophils to native pentameric CRP led to a dose- and time-dependent loss of C5aR expression on neutrophils, whereas the monomeric form of CRP, as well as various other inflammatory mediators, failed to significantly alter C5aR levels on neutrophils. A circulating form of C5aR (cC5aR) was detected in serum by immunoblotting and a flow-based capture assay, suggestive of an intact C5aR molecule. Levels of cC5aR were significantly enhanced during septic shock, with serum levels directly correlating with lethality. The data suggest that septic shock in humans is associated with extensive complement activation, CRP-dependent loss of C5aR on neutrophils, and appearance of cC5aR in serum, which correlated with a poor outcome. Therefore, cC5aR may represent a new sepsis marker to be considered in tailoring individualized immune-modulating therapy.


Asunto(s)
Neutrófilos/inmunología , Neutrófilos/metabolismo , Receptores de Complemento/sangre , Choque Séptico/sangre , Choque Séptico/inmunología , Adulto , Anciano , Animales , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Sprague-Dawley , Receptor de Anafilatoxina C5a , Receptores de Complemento/antagonistas & inhibidores , Choque Séptico/mortalidad , Sobrevida
7.
Mediators Inflamm ; 2015: 463950, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26556956

RESUMEN

Severe tissue trauma-induced systemic inflammation is often accompanied by evident or occult blood-organ barrier dysfunctions, frequently leading to multiple organ dysfunction. However, it is unknown whether specific barrier molecules are shed into the circulation early after trauma as potential indicators of an initial barrier dysfunction. The release of the barrier molecule junctional adhesion molecule-1 (JAM-1) was investigated in plasma of C57BL/6 mice 2 h after experimental mono- and polytrauma as well as in polytrauma patients (ISS ≥ 18) during a 10-day period. Correlation analyses were performed to indicate a linkage between JAM-1 plasma concentrations and organ failure. JAM-1 was systemically detected after experimental trauma in mice with blunt chest trauma as a driving force. Accordingly, JAM-1 was reduced in lung tissue after pulmonary contusion and JAM-1 plasma levels significantly correlated with increased protein levels in the bronchoalveolar lavage as a sign for alveolocapillary barrier dysfunction. Furthermore, JAM-1 was markedly released into the plasma of polytrauma patients as early as 4 h after the trauma insult and significantly correlated with severity of disease and organ dysfunction (APACHE II and SOFA score). The data support an early injury- and time-dependent appearance of the barrier molecule JAM-1 in the circulation indicative of a commencing trauma-induced barrier dysfunction.


Asunto(s)
Moléculas de Adhesión Celular/sangre , Traumatismo Múltiple/sangre , Receptores de Superficie Celular/sangre , APACHE , Animales , Líquido del Lavado Bronquioalveolar/química , Humanos , Ratones , Ratones Endogámicos C57BL , Índice de Severidad de la Enfermedad
8.
J Immunol ; 188(6): 2858-65, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22308306

RESUMEN

Severe tissue injury results in early activation of serine protease systems including the coagulation and complement cascade. In this context, little is known about factor VII-activating protease (FSAP), which is activated by substances released from damaged cells such as histones and nucleosomes. Therefore, we have measured FSAP activation in trauma patients and have identified novel FSAP substrates in human plasma. Mass spectrometry-based methods were used to identify FSAP binding proteins in plasma. Anaphylatoxin generation was measured by ELISA, Western blotting, protein sequencing, and chemotaxis assays. Plasma samples from trauma patients were analyzed for FSAP Ag and activity, nucleosomes, C5a, and C3a. Among others, we found complement components C3 and C5 in FSAP coimmunoprecipitates. C3 and C5 were cleaved by FSAP in a dose- and time-dependent manner generating functional C3a and C5a anaphylatoxins. Activation of endogenous FSAP in plasma led to increased C5a generation, but this was not the case in plasma of a homozygous carrier of Marburg I single nucleotide polymorphism with lower FSAP activity. In multiple trauma patients there was a large increase in circulating FSAP activity and nucleosomes immediately after the injury. A high correlation between FSAP activity and C5a was found. These data suggest that activation of FSAP by tissue injury triggers anaphylatoxin generation and thereby modulates the posttraumatic inflammatory response in vivo. A strong link between C5a, nucleosomes, and FSAP activity indicates that this new principle might be important in the regulation of inflammation.


Asunto(s)
Complemento C5a/inmunología , Traumatismo Múltiple/inmunología , Serina Endopeptidasas/inmunología , Adulto , Anciano , Western Blotting , Complemento C5a/metabolismo , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Inmunoprecipitación , Masculino , Espectrometría de Masas , Persona de Mediana Edad , Traumatismo Múltiple/sangre , Serina Endopeptidasas/sangre , Adulto Joven
9.
Adv Exp Med Biol ; 946: 135-46, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-21948366

RESUMEN

In response to severe tissue trauma, several "molecular danger" sensing and signaling pathways are activated, especially the complement and the apoptosis cascade. Although possible crossroads between both systems have been proposed, little is known about the underlying molecular interactions. In this study a new interaction interface is presented for C3a and C5a generation by the pro-apoptotic factor granzyme B. In vitro incubation of the central human complement components C3 and C5 with the serine protease granzyme B resulted in a concentration-dependent production of the anaphylatoxins C3a and C5a. The so generated anaphylatoxin C5a was chemotactic active for isolated human neutrophils. In a translational approach, intracellular granzyme B concentration in leukocytes was determined early after severe tissue trauma. In comparison to healthy volunteers, multiple injured patients (less than one hour after trauma, Injury Severity Score > 18, n = 5) presented a significant increase in granzmye B levels in neutrophils and lymphocytes. Thus, tissue trauma is associated with early activation of both, the complement and apoptosis system. The present data suggest a new form of interaction between the complement and the apoptosis system on the level of granzyme B that is capable to generate C3a and C5a independently of the established complement proteases.


Asunto(s)
Apoptosis/inmunología , Proteínas del Sistema Complemento/inmunología , Granzimas/inmunología , Traumatismo Múltiple/inmunología , Transducción de Señal/inmunología , Humanos
10.
Shock ; 51(1): 78-87, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-29461464

RESUMEN

Trauma-induced hemorrhagic shock (HS) plays a decisive role in the development of immune, coagulation, and organ dysfunction often resulting in a poor clinical outcome. Imbalanced complement activation is intricately associated with the molecular danger response and organ damage after HS. Thus, inhibition of the central complement component C3 as turnstile of both inflammation and coagulation is hypothesized as a rational strategy to improve the clinical course after HS.Applying intensive care conditions, anaesthetized, monitored, and protectively ventilated nonhuman primates (NHP; cynomolgus monkeys) received a pressure-controlled severe HS (60 min at mean arterial pressure 30 mmHg) with subsequent volume resuscitation. Thirty minutes after HS, animals were randomly treated with either an analog of the C3 inhibitor compstatin (i.e., Cp40) in saline (n = 4) or with saline alone (n = 4). The observation period lasted 300 min after induction of HS.We observed improved kidney function in compstatin Cp40-treated animals after HS as determined by improved urine output, reduced damage markers and a tendency of less histopathological signs of acute kidney injury. Sham-treated animals revealed classical signs of mucosal edema, especially in the ileum and colon reflected by worsened microscopic intestinal injury scores. In contrast, Cp40-treated HS animals exhibited only minor signs of organ edema and significantly less intestinal damage. Furthermore, early systemic inflammation and coagulation dysfunction were both ameliorated by Cp40.The data suggest that therapeutic inhibition of C3 is capable to significantly improve immune, coagulation, and organ function and to preserve organ-barrier integrity early after traumatic HS. C3-targeted complement inhibition may therefore reflect a promising therapeutic strategy in fighting fatal consequences of HS.


Asunto(s)
Inactivadores del Complemento , Péptidos Cíclicos , Choque Hemorrágico , Animales , Masculino , Inactivadores del Complemento/farmacología , Macaca fascicularis , Péptidos Cíclicos/farmacología , Choque Hemorrágico/sangre , Choque Hemorrágico/patología , Choque Hemorrágico/prevención & control
11.
J Crit Care ; 44: 229-237, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29175047

RESUMEN

Polytrauma (PT) is frequently associated with hemorrhagic shock (HS), which increases morbidity and mortality. Although various aspects of HS have been addressed in PT patients, the impact of an additional HS is largely unknown regarding the development of multiple organ dysfunction associated with disturbed glycocalyx and barrier function early after trauma. A prospective, longitudinal, mono-centered, observational study enrolling severely injured patients (Injury Severity Score, ISS=38.1±2.6) served for an in-depth analysis of blood (drawn on days 0, 1, 2, 3 and 5) and clinical data (up to 21days) of 30 patients who were then stratified into PT with and without HS. HS significantly enhanced signs of acute organ injury, assessed by increased serum concentrations of novel damage markers. Moreover, indicators of glycocalyx and tight-junction dysfunction were found in PT patients all of which were significantly enhanced in co-presence of HS. These markers revealed multiple significant correlations with specific barrier, fluid-balance, coagulation, inflammation, and clinical-outcome parameters. Strikingly, mucosa fragments, which affected clotting, could be detected in serum after PT/HS. The results point to HS as a main driver for glycocalyx and barrier breakdown and suggest novel tools for the monitoring of organ dysfunction in the early course after PT.


Asunto(s)
Biomarcadores/sangre , Glicocálix/metabolismo , Traumatismo Múltiple , Choque Hemorrágico/mortalidad , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Servicio de Urgencia en Hospital , Femenino , Alemania/epidemiología , Humanos , Puntaje de Gravedad del Traumatismo , Estudios Longitudinales , Masculino , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Estudios Prospectivos , Choque Hemorrágico/sangre , Adulto Joven
12.
Shock ; 49(2): 154-163, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28614141

RESUMEN

Hemorrhagic shock (HS) after tissue trauma increases the complication and mortality rate of polytrauma (PT) patients. Although several murine trauma models have been introduced, there is a lack of knowledge about the exact impact of an additional HS. We hypothesized that HS significantly contributes to organ injury, which can be reliably monitored by detection of specific organ damage markers. Therefore we established a novel clinically relevant PT plus HS model in C57BL/6 mice which were randomly assigned to control, HS, PT, or PT+HS procedure (n = 8 per group). For induction of PT, anesthetized animals received a blunt chest trauma, head injury, femur fracture, and soft tissue injury. HS was induced by pressure-controlled blood drawing (mean arterial blood pressure of 30 mmHg for 60 min) and mice then resuscitated with ionosterile (4 × volume drawn), monitored, and killed for blood and organ harvesting 4 h after injury. After HS and resuscitation, PT+HS mice required earlier and overall more catecholamine support than HS animals to keep their mean arterial blood pressure. HS significantly contributed to the systemic release of interleukin-6 and high mobility group box 1 protein. Furthermore, the histological lung injury score, pulmonary edema, neutrophil influx, and plasma clara cell protein 16 were all significantly enhanced in PT animals in the presence of an additional HS. Although early morphological changes were minor, HS also contributed functionally to remote acute kidney injury but not to early liver damage. Moreover, PT-induced systemic endothelial injury, as determined by plasma syndecan-1 levels, was significantly aggravated by an additional HS. These results indicate that HS adds to the systemic inflammatory reaction early after PT. Within hours after PT, HS seems to aggravate pulmonary damage and to worsen renal and endothelial function which might overall contribute to the development of early multiple organ dysfunction.


Asunto(s)
Traumatismo Múltiple/sangre , Traumatismo Múltiple/fisiopatología , Choque Hemorrágico/sangre , Choque Hemorrágico/fisiopatología , Animales , Lavado Broncoalveolar , Creatinina/sangre , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Proteína HMGB1/metabolismo , Interleucina-6/sangre , Riñón/metabolismo , Ratones , Ratones Endogámicos C57BL , Traumatismo Múltiple/metabolismo , Peroxidasa/metabolismo , Distribución Aleatoria , Choque Hemorrágico/metabolismo
13.
Front Immunol ; 8: 15, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28144242

RESUMEN

The complement system is a major pillar of the humoral innate immune system. As a first line of defense against pathogens, it mediates early inflammatory response and links different branches of humoral and cellular immunity. Disorders affecting the exocrine pancreas, such as acute pancreatitis, potentially lead to a life-threatening systemic inflammatory response with aberrant activation of complement and coagulation cascades. Pancreatic proteases can activate key effectors of the complement system, which in turn drive local and systemic inflammation. Beyond that, the extent of pancreas-complement interaction covers complex pro- and anti-inflammatory mechanisms, which to this day remain to be fully elucidated. This review provides a comprehensive overview of the pathophysiological role of complement in diseases of the exocrine pancreas, based on existing experimental and clinical data. Participation of complement in acute and chronic pancreatitis is addressed, as well as its role in tumor immunology. Therapeutic strategies targeting complement in these diseases have long been proposed but have not yet arrived in the clinical setting.

14.
Shock ; 46(4): 398-404, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-26926005

RESUMEN

Septic shock-related kidney failure is characterized by almost normal morphological appearance upon pathological examination. Endothelial barrier disrupture has been suggested to be of crucial importance for septic shock-induced organ dysfunction. Therefore, in murine resuscitated cecal ligation and puncture (CLP)-induced septic shock, we tested the hypothesis whether there is a direct relationship between the kidney endothelial barrier injury and renal dysfunction. Anesthetized mice underwent CLP, and 15 h later, were anesthetized again and surgically instrumented for a 5-h period of intensive care comprising lung-protective mechanical ventilation, fluid resuscitation, continuous i.v. norepinephrine to maintain target hemodynamics, and measurement of creatinine clearance (CrCl). Animals were stratified according to low or high CrCl. Nitrotyrosine formation, expression of the inducible isoform of the nitric oxide synthase, and blood cytokine (tumor necrosis factor, interleukin-6, interleukin-10) and chemokine (monocyte chemoattractant protein-1, keratinocyte-derived chemokine) levels were significantly higher in animals with low CrCl. When plotted against CrCl and neutrophil gelatinase-associated lipocalin levels, extravascular albumin accumulation, and tissue expression of the vascular endothelial growth factor and angiopoietin-1 showed significant mathematical relationships related to kidney (dys)function. Preservation of the constitutive expression of the hydrogen sulfide producing enzyme cystathione-γ-lyase was associated with maintenance of organ function. The direct quantitative relation between microvascular leakage and kidney (dys)function may provide a missing link between near-normal tissue morphology and septic shock-related renal failure, thus further highlighting the important role of vascular integrity in septic shock-related renal failure.


Asunto(s)
Riñón/metabolismo , Choque Séptico/metabolismo , Choque Séptico/fisiopatología , Angiopoyetina 1/metabolismo , Animales , Quimiocina CCL2/metabolismo , Quimiocinas/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Hemodinámica/fisiología , Inmunohistoquímica , Riñón/fisiopatología , Ratones , Modelos Teóricos , Óxido Nítrico Sintasa/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
15.
Eur J Med Res ; 20: 70, 2015 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-26303896

RESUMEN

BACKGROUND: After severe blunt chest trauma, the development of an acute lung injury (ALI) is often associated with severe or even lethal complications. Especially in multiple injured patients after blunt chest trauma ALI/ARDS [acute respiratory distress syndrome (ARDS)] is frequent. However, in the initial posttraumatic phase, inflammatory clinical signs are often not apparent and underlying changes in gene-expression profile are unknown. METHODS: Therefore, inflammation in lung tissue following blunt chest trauma was characterized in a well-defined bilateral lung injury model. Using DNA microarrays representing 9240 genes, the temporal sequence of blunt chest trauma-induced gene-expression patterns in lung tissue was examined. RESULTS: The results suggest an activation of a highly complex transcriptional program in response to chest trauma. Chest trauma led to elevated expression levels of inflammatory and coagulatory proteins (such as TNFα receptor, IL-1α, IL-1ß, C3, NF-κB and plasminogen activator). However, upregulation of proteins was found, usually incoherent of exerting effects in blunt thoracic trauma (pendrin, resistin, metallothionein and glucocorticoid-induced leucine zipper). Furthermore, significant downregulation was observed as early as 10 min after trauma for cytokines and complement factors (LCR-1, C4) as well as for intracellular signaling molecules (inhibitory protein phosphatase) and ion-channels (voltage-dependent Ca(2+) channel). CONCLUSIONS: Taken together, the provided global perspective of the inflammatory response following blunt chest trauma could provide a molecular framework for future research in trauma pathophysiology.


Asunto(s)
Neumonía/etiología , Traumatismos Torácicos/complicaciones , Heridas no Penetrantes/complicaciones , Proteínas del Sistema Complemento/metabolismo , Perfilación de la Expresión Génica , Hemo-Oxigenasa 1/metabolismo , Humanos , Canales Iónicos/metabolismo , FN-kappa B/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Neumonía/genética
16.
PLoS One ; 9(1): e87294, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24489892

RESUMEN

The family of vertebrate Mef2 transcription factors is comprised of four members named Mef2a, Mef2b, Mef2c, and Mef2d. These transcription factors are regulators of the myogenic programs with crucial roles in development of skeletal, cardiac and smooth muscle cells. Mef2a and Mef2c are essential for cardiac development in mice. In Xenopus, mef2c and mef2d but not mef2a were recently shown to be expressed during cardiogenesis. We here investigated the function of Mef2c and Mef2d during Xenopus laevis cardiogenesis. Knocking down either gene by corresponding antisense morpholino oligonucleotides led to profound heart defects including morphological abnormalities, pericardial edema, and brachycardia. Marker gene expression analyses and rescue experiments revealed that (i) both genes are required for proper cardiac gene expression, (ii) Mef2d can compensate for the loss of Mef2c but not vice versa, and (iii) the γ domain of Mef2c is required for early cardiac development. Taken together, our data provide novel insights into the function of Mef2 during cardiogenesis, highlight evolutionary differences between species and might have an impact on attempts of direct reprogramming.


Asunto(s)
Factores de Transcripción MEF2/fisiología , Organogénesis/genética , Animales , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Corazón/embriología , Cardiopatías Congénitas/genética , Factores de Transcripción MEF2/metabolismo , Estructura Terciaria de Proteína , Xenopus laevis
17.
J Trauma Acute Care Surg ; 74(3): 792-800, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23425737

RESUMEN

BACKGROUND: Direct acute lung injury (ALI) is still associated with a high mortality, whereas the underlying pathomechanisms are not yet fully understood. In this regard, epithelial cell death in the lungs has been attributed an important role in the pathogenesis of this clinical entity. Based on this background here, we hypothesized that signaling through Fas and tumor necrosis factor receptor 1 (TNFR-1) is involved in mediating apoptosis and inflammation in chest trauma induced septic ALI. METHODS: Male C57BL/6 mice (wild-type [WT]), male mutant mice expressing nonfunctional Fas receptor (B6.MRL-Faslpr/J [lpr]) (lpr) and male TNFR-1-deficient mice (TNFR-1(-/-)) were subjected to a model of direct ALI consisting of blunt chest trauma followed by cecal ligation and puncture.Cytokine/chemokine concentrations of plasma, bronchoalveolar lavage (BAL) fluids, and lung tissue were investigated as well as BAL protein and lung myeloperoxidase. Lung histology was assessed; lung caspase 3, TUNEL-positive cells, and apoptotic polymorphonuclear neutrophil were measured, followed by a survival study. RESULTS: Cytokine/chemokine levels in plasma, BAL, and lung tissue were markedly increased in WT animals following ALI, whereas lpr and TNFR-1((-/-) showed significantly decreased levels. BAL protein levels were substantially elevated following ALI, but lpr animals presented markedly diminished protein levels compared with WT and TNFR-1(-/-) animals. Lung myeloperoxidase level was only increased 12 hours after ALI in WT animals, whereas lung myeloperoxidase levels in lpr and TNFR-1(-/-) animals were not increased compared with sham. Lung histology revealed beneficial effects in lpr and TNFR-1(-/-). Lung active caspase 3 after ALI was substantially decreased in lpr and TNFR-1(-/-) mice compared with WT. Interestingly, an early but not persisting survival benefit was observed in lpr and TNFR-1 animals(-/-). CONCLUSION: Pathomechanistically, Fas and TNFR-1 signaling contributed to the apoptotic and inflammatory response in a clinically relevant double-hit model of trauma-induced septic ALI. Moreover, this was associated with a temporary survival benefit.


Asunto(s)
Lesión Pulmonar Aguda/etiología , Apoptosis , Líquido del Lavado Bronquioalveolar/química , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Sepsis/complicaciones , Traumatismos Torácicos/complicaciones , Receptor fas/metabolismo , Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/patología , Animales , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Etiquetado Corte-Fin in Situ , Masculino , Ratones , Ratones Endogámicos C57BL , Sepsis/metabolismo , Sepsis/patología , Transducción de Señal , Traumatismos Torácicos/metabolismo , Traumatismos Torácicos/patología
18.
J Trauma Acute Care Surg ; 74(2): 489-98, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23354243

RESUMEN

BACKGROUND: The exact alterations of the immune system after polytrauma leading to sepsis and multiple-organ failure are poorly understood. Thus, the early local and systemic inflammatory and apoptotic response was characterized in a new polytrauma model and compared with the alterations seen after single or combined injuries. METHODS: Anesthetized C57BL/6 mice were subjected to either blunt bilateral chest trauma (Tx), closed head injury, right femur fracture including contralateral soft tissue injury, or a combination of injuries (PTx). After 2 hours or 6 hours, animals were sacrificed, and the systemic as well as the local pulmonary immune response (bronchoalveolar lavage [BAL]/plasma cytokines, lung myeloperoxidase [MPO] activity, and alveolocapillary barrier dysfunction) were evaluated along with lung/brain apoptosis (lung caspase 3 Western blotting, immunohistochemistry, and polymorphonuclear leukocytes [PMN] Annexin V). RESULTS: Hemoglobin, PO2 saturation, and pH did not differ between the experimental groups. Local BAL cytokines/chemokines were significantly increased in almost all groups, which included Tx. There was no further enhancement of this local inflammatory response in the lungs in case of PTx. At 2 hours, all groups except sham and closed head injury alone revealed an increased activity of lung MPO. However, 6 hours after injury, lung MPO remained increased only in the PTx group. Increased BAL protein levels were found, reflecting enhanced lung leakage in all groups with Tx 6 hours after trauma. Only after PTx was neutrophil apoptosis significantly decreased, whereas lung caspase 3 and plasma interleukin 6/keratinocyte chemoattractant (KC) were substantially increased. CONCLUSION: The combination of different injuries leads to an earlier systemic inflammatory response when compared with the single insults. Interestingly, only after PTx but not after single or double hits was lung apoptosis increased, and PMN apoptosis was decreased along with a prolonged presence of neutrophils in the lungs, which may therefore represent a possible pathomechanism for lung injury after polytrauma.


Asunto(s)
Apoptosis/fisiología , Inflamación/etiología , Traumatismo Múltiple/complicaciones , Heridas y Lesiones/complicaciones , Animales , Western Blotting , Encéfalo/inmunología , Encéfalo/fisiopatología , Líquido del Lavado Bronquioalveolar/química , Caspasa 3/metabolismo , Quimiocinas/análisis , Quimiocinas/sangre , Citocinas/análisis , Citocinas/sangre , Citometría de Flujo , Hemoglobinas/análisis , Inflamación/sangre , Inflamación/inmunología , Inflamación/fisiopatología , Pulmón/química , Pulmón/inmunología , Pulmón/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Traumatismo Múltiple/sangre , Traumatismo Múltiple/inmunología , Traumatismo Múltiple/fisiopatología , Peroxidasa/metabolismo , Heridas y Lesiones/sangre , Heridas y Lesiones/inmunología , Heridas y Lesiones/fisiopatología
19.
Shock ; 39(1): 19-27, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23247118

RESUMEN

Activation of Fas signaling is a potentially important pathophysiological mechanism in the development of septic acute lung injury (ALI). However, so far the optimal targets within this signaling cascade remain elusive. Thus, we tested the hypothesis that in vivo gene silencing of Fas, Fas-associated via death domain (FADD), or caspase 3 by intratracheal administration of small interfering RNA would ameliorate ALI in a clinically relevant double-hit mouse model of trauma induced septic lung injury. Male C57Bl/6 mice received small interfering (Fas, FADD, caspase 3) or control RNA 24 h before and 12 h after blunt chest trauma or sham procedures. Polymicrobial sepsis was induced by cecal ligation and puncture 24 h after chest trauma. Twelve or 24 h later, lung tissue, plasma, and bronchoalveolar lavage fluid were harvested. During ALI, lung apoptosis (active caspase 3 Western blotting, TUNEL staining) was substantially increased when compared with sham. Silencing of caspase 3 or FADD both markedly reduced pulmonary apoptosis. Fas- and FADD-small interfering RNA administration substantially decreased lung cytokine concentration, whereas caspase 3 silencing did not reduce lung inflammation. In addition, Fas silencing markedly decreased lung neutrophil infiltration. Interestingly, only in response to caspase 3 silencing, ALI-induced lung epithelial barrier dysfunction was substantially improved, and histological appearance was beneficially affected. Taken together, downstream inhibition of lung apoptosis via caspase 3 silencing proved to be superior in mitigating ALI when compared with upstream inhibition of apoptosis via Fas or FADD silencing, even in the presence of additional anti-inflammatory effects. This indicates a major pathophysiological role of lung apoptosis and suggests the importance of other than Fas-driven apoptotic pathways in trauma-induced septic ALI.


Asunto(s)
Lesión Pulmonar Aguda/prevención & control , Caspasa 3/genética , Proteína de Dominio de Muerte Asociada a Fas/genética , Terapia Genética/métodos , Neumonía/prevención & control , Receptor fas/genética , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/patología , Animales , Apoptosis/genética , Caspasa 3/fisiología , Citocinas/sangre , Modelos Animales de Enfermedad , Proteína Ligando Fas/sangre , Proteína de Dominio de Muerte Asociada a Fas/fisiología , Silenciador del Gen , Interleucina-10/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Infiltración Neutrófila/genética , Neumonía/genética , Neumonía/patología , ARN Interferente Pequeño/genética , Transducción de Señal/genética , Heridas no Penetrantes/genética , Heridas no Penetrantes/patología , Receptor fas/fisiología
20.
Mol Immunol ; 50(1-2): 60-5, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22244896

RESUMEN

In response to severe tissue trauma several danger sensing and signalling cascades are activated, including the complement and the apoptosis systems. In polytrauma patients, both the early activation of the complement cascade with an excessive generation of the potent anaphylatoxin C5a and the induction of apoptosis have been shown to modulate the post-traumatic immune response. However, little is known about a direct interaction between the complement and apoptosis systems after severe tissue trauma. Therefore the focus of the present study was to elucidate the interplay between the central complement component C5 and the pro-apoptotic aspartic protease cathepsin D. In vivo, the cathepsin D plasma concentration of multiple injured patients was markedly increased when compared to healthy volunteers. In vitro incubation of C5 with cathepsin D resulted in a concentration- and time-dependent generation of C5a, which was inhibited by the aspartate protease inhibitor pepstatin A. Immunoblotting and sequencing analysis indicated that the C5 cleavage product represents the native form of human C5a, also exhibiting chemotactic activity for human neutrophils. In conclusion, these data show for the first time that cathepsin D is increased in plasma early after severe tissue injury. Furthermore, the results provide in vitro evidence of cleavage of C5 by an aspartic protease with subsequent generation of functional C5a, which represents a new path of complement activation.


Asunto(s)
Catepsina D/metabolismo , Complemento C5/metabolismo , Complemento C5a/metabolismo , Heridas y Lesiones/metabolismo , Biocatálisis/efectos de los fármacos , Western Blotting , Catepsina D/antagonistas & inhibidores , Catepsina D/sangre , Quimiotaxis de Leucocito , Activación de Complemento , Ensayo de Inmunoadsorción Enzimática , Humanos , Neutrófilos/citología , Neutrófilos/metabolismo , Pepstatinas/farmacología , Inhibidores de Proteasas/farmacología , Factores de Tiempo , Heridas y Lesiones/sangre , Heridas y Lesiones/enzimología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA