Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Shock ; 49(2): 154-163, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28614141

RESUMO

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.


Assuntos
Traumatismo Múltiplo/sangue , Traumatismo Múltiplo/fisiopatologia , Choque Hemorrágico/sangue , Choque Hemorrágico/fisiopatologia , Animais , Lavagem Broncoalveolar , Creatinina/sangue , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Proteína HMGB1/metabolismo , Interleucina-6/sangue , Rim/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Traumatismo Múltiplo/metabolismo , Peroxidase/metabolismo , Distribuição Aleatória , Choque Hemorrágico/metabolismo
2.
Shock ; 47(2): 225-235, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27405068

RESUMO

Pulmonary Fas activation is essential in the pathogenesis of the acute respiratory distress syndrome. It remains unclear whether Fas-induced lung injury is dependent on neutrophils or mainly triggered by epithelial cell apoptosis. The contribution of lung epithelial cells (LEC) and alveolar macrophages (AM) remains elusive.Mice were neutrophil reduced prior to intratracheal instillation of Fas-activating (Jo2) or isotype antibody for 6 or 18 h. LEC and AM were incubated with Jo2 and in the presence of nuclear factor kappa B, p-38 mitogen activated protein kinase (p38MAPK), or extracellular signal regulating kinase 1/2 (ERK1/2) inhibitors. Cytokines were assessed by cytometric bead array or ELISA. Apoptosis was quantified via active caspase-3 Western blotting and Terminal Deoxynucleotide Transferase dUTP Nick End Labeling (TUNEL). Lung injury was assessed by bronchoalveolar lavage fluid (BALF) protein concentration and lung histology.KC, IL-6, and MCP-1 were markedly increased in lung, plasma, and BALF 18 h after Jo2 in the presence of neutrophils; in neutrophil-reduced mice lungs, MCP-1, but not KC or IL-6, was even further enhanced. Six hours after Jo2, BALF protein was markedly increased only in the presence of neutrophils. Apoptosis remained unaffected by neutrophil reduction. AM released MCP-1 and underwent apoptosis at lower concentrations of Jo2 than LEC. Inhibition of p38MAPK significantly increased, while inhibition of ERK1/2 reduced AM and LEC apoptosis.In conclusion, neutrophils are a necessary component of Fas-induced lung damage, while not affecting lung apoptosis directly per se. LEC display higher resistance to Fas-triggered inflammation and apoptosis than AM.


Assuntos
Neutrófilos/metabolismo , Pneumonia/metabolismo , Receptor fas/metabolismo , Animais , Apoptose/fisiologia , Líquido da Lavagem Broncoalveolar , Quimiocina CCL2/metabolismo , Ensaio de Imunoadsorção Enzimática , Proteína Ligante Fas/metabolismo , Marcação In Situ das Extremidades Cortadas , Inflamação/metabolismo , Interleucina-6/metabolismo , Pulmão/citologia , Pulmão/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
3.
PLoS One ; 11(7): e0159417, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27437704

RESUMO

BACKGROUND: Severe blunt chest trauma is associated with high mortality. Sepsis represents a serious risk factor for mortality in acute respiratory distress syndrome (ARDS). In septic patients with ARDS complement activation products were found to be elevated in the plasma. In single models like LPS or trauma complement has been studied to some degree, however in clinically highly relevant double hit models such as the one used here little data is available. Here, we hypothesized that absence of C5 is correlated with a decreased inflammatory response in trauma induced septic acute lung injury. METHODS: 12 hrs after DH in mice the local and systemic cytokines and chemokines were quantified by multiplex bead array or ELISA, activated caspase-3 by western blot. Data were analyzed using one-way ANOVA followed by post-hoc Sidak's multiple comparison test (significance, p≤ 0.05). RESULTS: In lung tissue interleukin (IL)-6, monocyte chemo attractant protein-1 (MCP-1) and granulocyte-colony stimulating factor (G-CSF) was elevated in both C5-/- mice and wildtype littermates (wt), whereas caspase-3 was reduced in lungs after DH in C5-/- mice. Systemically, reduced keratinocyte-derived chemokine (KC) levels were observed after DH in C5-/- compared to wt mice. Locally, lung myeloperoxidase (MPO), protein, IL-6, MCP-1 and G-CSF in brochoalveolar lavage fluid (BALF) were elevated after DH in C5-/- compared to wt. CONCLUSIONS: In the complex but clinically relevant DH model the local and systemic inflammatory immune response features both, C5-dependent and C5-independent characteristics. Activation of caspase-3 in lung tissue after DH was C5-dependent whereas local inflammation in lung tissue was C5-independent.


Assuntos
Lesão Pulmonar Aguda/genética , Complemento C5/genética , Inflamação/genética , Síndrome do Desconforto Respiratório/genética , Sepse/genética , Lesão Pulmonar Aguda/patologia , Animais , Caspase 3/biossíntese , Quimiocina CCL2/biossíntese , Complemento C5/metabolismo , Fator Estimulador de Colônias de Granulócitos/biossíntese , Humanos , Inflamação/complicações , Inflamação/patologia , Interleucina-6/biossíntese , Camundongos , Síndrome do Desconforto Respiratório/patologia , Fatores de Risco , Sepse/sangue , Sepse/complicações , Sepse/patologia , Traumatismos Torácicos/genética , Traumatismos Torácicos/patologia , Ferimentos não Penetrantes/genética
4.
J Trauma Acute Care Surg ; 74(3): 792-800, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23425737

RESUMO

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.


Assuntos
Lesão Pulmonar Aguda/etiologia , Apoptose , Líquido da Lavagem Broncoalveolar/química , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Sepse/complicações , Traumatismos Torácicos/complicações , Receptor fas/metabolismo , Lesão Pulmonar Aguda/metabolismo , Lesão Pulmonar Aguda/patologia , Animais , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Marcação In Situ das Extremidades Cortadas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Sepse/metabolismo , Sepse/patologia , Transdução de Sinais , Traumatismos Torácicos/metabolismo , Traumatismos Torácicos/patologia
5.
Shock ; 39(1): 19-27, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23247118

RESUMO

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.


Assuntos
Lesão Pulmonar Aguda/prevenção & controle , Caspase 3/genética , Proteína de Domínio de Morte Associada a Fas/genética , Terapia Genética/métodos , Pneumonia/prevenção & controle , Receptor fas/genética , Lesão Pulmonar Aguda/genética , Lesão Pulmonar Aguda/patologia , Animais , Apoptose/genética , Caspase 3/fisiologia , Citocinas/sangue , Modelos Animais de Doenças , Proteína Ligante Fas/sangue , Proteína de Domínio de Morte Associada a Fas/fisiologia , Inativação Gênica , Interleucina-10/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Infiltração de Neutrófilos/genética , Pneumonia/genética , Pneumonia/patologia , RNA Interferente Pequeno/genética , Transdução de Sinais/genética , Ferimentos não Penetrantes/genética , Ferimentos não Penetrantes/patologia , Receptor fas/fisiologia
6.
Shock ; 38(1): 98-106, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22552016

RESUMO

More than 50% of severely injured patients have chest trauma. Second insults frequently result in acute lung injury (ALI), with sepsis being the main underlying condition. We aimed to develop a standardized, reproducible, and clinically relevant double-hit mouse model of ALI induced by chest trauma and polymicrobial sepsis and to investigate the pathophysiologic role of activated neutrophils. Lung contusion was applied to C57Bl/6 mice via a focused blast wave. Twenty-four hours later, sepsis was induced by cecal ligation and puncture. For polymorphonuclear leukocyte (PMN) depletion, animals received intravenous injections of PMN-depleting antibody. In response to blunt chest trauma followed by sepsis as well as after sepsis alone, a significant local and systemic inflammatory response with increased cytokine/chemokine levels in lung and plasma was observed. In contrast, lung apoptosis was markedly elevated only after a double hit. Intra-alveolar neutrophils and total bronchoalveolar lavage protein concentrations were markedly increased following isolated chest trauma or the combined insult, but not after sepsis alone. Lung myeloperoxidase activity was enhanced only in response to the double hit accompanied by histological disruption of the alveolar architecture, lung congestion, and marked cellular infiltrates. Neutrophil depletion significantly diminished lung interleukin 1ß and interleukin 6 concentrations and reduced the degree of septic ALI. Here we have established a novel and highly reproducible mouse model of chest trauma-induced septic ALI characterizing a clinical relevant double-hit scenario. In particular, the depletion of neutrophils substantially mitigated the extent of lung injury, indicating a pathomechanistic role for neutrophils in chest trauma-induced septic ALI.


Assuntos
Lesão Pulmonar Aguda/etiologia , Ativação de Neutrófilo/imunologia , Neutrófilos/imunologia , Sepse/complicações , Traumatismos Torácicos/complicações , Lesão Pulmonar Aguda/imunologia , Lesão Pulmonar Aguda/patologia , Animais , Apoptose/imunologia , Líquido da Lavagem Broncoalveolar/química , Dióxido de Carbono/sangue , Caspases/metabolismo , Quimiocinas/sangue , Citocinas/sangue , Modelos Animais de Doenças , Pulmão/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Oxigênio/sangue , Pressão Parcial , Sepse/imunologia , Síndrome de Resposta Inflamatória Sistêmica/etiologia , Síndrome de Resposta Inflamatória Sistêmica/imunologia , Ferimentos não Penetrantes/complicações , Receptor fas/sangue
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA