Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
2.
J Cell Biol ; 222(2)2023 02 06.
Article in English | MEDLINE | ID: mdl-36459066

ABSTRACT

Progressive accrual of senescent cells in aging and chronic diseases is associated with detrimental effects in tissue homeostasis. We found that senescent fibroblasts and epithelia were not only refractory to macrophage-mediated engulfment and removal, but they also paralyzed the ability of macrophages to remove bystander apoptotic corpses. Senescent cell-mediated efferocytosis suppression (SCES) was independent of the senescence-associated secretory phenotype (SASP) but instead required direct contact between macrophages and senescent cells. SCES involved augmented senescent cell expression of CD47 coinciding with increased CD47-modifying enzymes QPCT/L. SCES was reversible by interfering with the SIRPα-CD47-SHP-1 axis or QPCT/L activity. While CD47 expression increased in human and mouse senescent cells in vitro and in vivo, another ITIM-containing protein, CD24, contributed to SCES specifically in human epithelial senescent cells where it compensated for genetic deficiency in CD47. Thus, CD47 and CD24 link the pathogenic effects of senescent cells to homeostatic macrophage functions, such as efferocytosis, which we hypothesize must occur efficiently to maintain tissue homeostasis.


Subject(s)
Apoptosis , CD47 Antigen , Macrophages , Senescence-Associated Secretory Phenotype , Animals , Humans , Mice , Aminoacyltransferases/metabolism , CD24 Antigen/metabolism , CD47 Antigen/genetics , CD47 Antigen/metabolism , Macrophages/cytology , Up-Regulation
3.
Br J Pharmacol ; 178(14): 2863-2879, 2021 07.
Article in English | MEDLINE | ID: mdl-32880897

ABSTRACT

Physical trauma represents a major global burden. The trauma-induced response, including activation of the innate immune system, strives for regeneration but can also lead to post-traumatic complications. The complement cascade is rapidly activated by damaged tissue, hypoxia, exogenous proteases and others. Activated complement can sense, mark and clear both damaged tissue and pathogens. However, excessive and insufficient activation of complement can result in a dysfunctional immune and organ response. Similar to acute coagulopathy, complementopathy can develop with enhanced anaphylatoxin generation and an impairment of complement effector functions. Various remote organ effects are induced or modulated by complement activation. Frequently, established trauma treatments are double-edged. On one hand, they help stabilising haemodynamics and oxygen supply as well as injured organs and on the other hand, they also drive complement activation. Immunomodulatory approaches aim to reset trauma-induced disbalance of complement activation and thus may change surgical trauma management procedures to improve outcome. LINKED ARTICLES: This article is part of a themed issue on Canonical and non-canonical functions of the complement system in health and disease. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.14/issuetoc.


Subject(s)
Complement System Proteins , Immune System , Humans , Hypoxia
4.
Front Immunol ; 11: 1789, 2020.
Article in English | MEDLINE | ID: mdl-32983087

ABSTRACT

Initially underestimated as platelet dust, extracellular vesicles are continuously gaining interest in the field of inflammation. Various studies addressing inflammatory diseases have shown that microvesicles (MVs) originating from different cell types are systemic transport vehicles carrying distinct cargoes to modulate immune responses. In this study, we focused on the clinical setting of multiple trauma, which is characterized by activation and dysfunction of both, the fluid-phase and the cellular component of innate immunity. Given the sensitivity of neutrophils for the complement anaphylatoxin C5a, we hypothesized that increased C5a production induces alterations in MV shedding of neutrophils resulting in neutrophil dysfunction that fuels posttraumatic inflammation. In a mono-centered prospective clinical study with polytraumatized patients, we found significantly increased granulocyte-derived MVs containing the C5a receptor (C5aR1, CD88) on their surface. This finding was accompanied by a concomitant loss of C5aR1 on granulocytes indicative of an impaired cellular chemotactic and pro-inflammatory neutrophil functions. Furthermore, in vitro exposure of human neutrophils (from healthy volunteers) to C5a significantly increased MV shedding and C5aR1 loss on neutrophils, which could be blocked using the C5aR1 antagonist PMX53. Mechanistic analyses revealed that the interaction between C5aR1 signaling and the small GTPase Arf6 acts as a molecular switch for MV shedding. When neutrophil derived, C5a-induced MV were exposed to a complex ex vivo whole blood model significant pro-inflammatory properties (NADPH activity, ROS and MPO generation) of the MVs became evident. C5a-induced MVs activated resting neutrophils and significantly induced IL-6 secretion. These data suggest a novel role of the C5a-C5aR1 axis: C5a-induced MV shedding from neutrophils results in decreased C5aR1 surface expression on the one hand, on the other hand it leads to profound inflammatory signals which likely are both key drivers of the neutrophil dysfunction which is regularly observed in patients suffering from multiple traumatic injuries.


Subject(s)
Cell-Derived Microparticles/immunology , Complement C5a/metabolism , Immunity, Innate , Inflammation Mediators/metabolism , Multiple Trauma/immunology , Neutrophils/immunology , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/metabolism , Adult , Case-Control Studies , Cell-Derived Microparticles/metabolism , Female , Humans , Injury Severity Score , Interleukin-6/metabolism , Kinetics , Male , Middle Aged , Multiple Trauma/blood , Multiple Trauma/diagnosis , NADP/metabolism , Neutrophils/metabolism , Peroxidase/metabolism , Prospective Studies , Reactive Oxygen Species/metabolism , Receptor, Anaphylatoxin C5a/metabolism , Signal Transduction , Young Adult
5.
Scand J Immunol ; 91(2): e12837, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31622512

ABSTRACT

After severe trauma, the resulting excessive inflammatory response is countered by compensatory anti-inflammatory mechanisms. The systemic inflammatory response to trauma enhanced by inappropriately timed surgical second hits may be detrimental for the patient. On the other hand, overwhelming anti-inflammatory mechanisms may put patients at increased risk from secondary local and systemic infections. The ensuing sepsis and organ dysfunction due to immune dysregulation remain the leading causes of death after injury. To date, there are no clinically applicable techniques to monitor the pro-/anti-inflammatory immune status of the patients and the remaining ability to react to microbial stimuli. Therefore, in the present study, we used a highly standardized and easy-to-use system to draw peripheral whole blood from polytraumatized patients (ISS ≥ 32, n = 7) and to challenge it with bacterial lipopolysaccharide. Secreted cytokines were compared with those in samples from healthy volunteers. We observed a significant decrease in the release of monocyte-derived mediators. Surprisingly, we detected stable or even increased concentrations of cytokines related to T cell maturation and function. For clinical practicability, we reduced the incubation time before supernatants were collected. Even after an abbreviated stimulation period, a stable release of almost all analysed parameters in patient blood could be detected. In conclusion, the data are indicative of a clinically well-applicable approach to monitor the immune status in severely injured patients in a short time. This may be used to optimize the timing of necessary surgical interventions to avoid a boost of proinflammation and reduce risk of secondary infections.


Subject(s)
Monitoring, Immunologic/methods , Multiple Trauma/diagnosis , Adult , Cells, Cultured , Disease Progression , Female , Humans , Lipopolysaccharides/immunology , Male , Middle Aged , Pilot Projects
6.
Mediators Inflamm ; 2018: 2052356, 2018.
Article in English | MEDLINE | ID: mdl-30002598

ABSTRACT

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.


Subject(s)
Complement C5a/pharmacology , Membrane Potentials/drug effects , Neutrophils/drug effects , Neutrophils/metabolism , Shock, Hemorrhagic/metabolism , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Electrophysiology , Flow Cytometry , Humans , Hydrogen-Ion Concentration , Male , Swine
7.
PLoS One ; 12(10): e0187327, 2017.
Article in English | MEDLINE | ID: mdl-29084268

ABSTRACT

Evidence is emerging that systemic inflammation after trauma drives structural and functional impairment of cardiomyocytes and leads to cardiac dysfunction, thus worsening the outcome of polytrauma patients. This study investigates the structural and molecular changes in heart tissue 4 h after multiple injuries with additional hemorrhagic shock using a clinically relevant rodent model of polytrauma. We determined mediators of systemic inflammation (keratinocyte chemoattractant, macrophage chemotactic protein 1), activated complement component C3a and cardiac troponin I in plasma and assessed histological specimen of the mouse heart via standard histomorphology and immunohistochemistry for cellular and subcellular damage and ongoing apoptosis. Further we investigated spatial and quantitative changes of connexin 43 by immunohistochemistry and western blotting. Our results show significantly increased plasma levels of both keratinocyte chemoattractant and cardiac troponin I 4 h after polytrauma and 2 h after induction of hypovolemia. Although we could not detect any morphological changes, immunohistochemical evaluation showed increased level of tissue high-mobility group box 1, which is both a damage-associated molecule and actively released as a danger response signal. Additionally, there was marked lateralization of the cardiac gap-junction protein connexin 43 following combined polytrauma and hemorrhagic shock. These results demonstrate a molecular manifestation of remote injury of cardiac muscle cells in the early phase after polytrauma and hemorrhagic shock with marked disruption of the cardiac gap junction. This disruption of an important component of the electrical conduction system of the heart may lead to arrhythmia and consequently to cardiac dysfunction.


Subject(s)
Heart/physiopathology , Multiple Trauma/pathology , Shock, Hemorrhagic/pathology , Humans
8.
FASEB J ; 31(9): 4129-4139, 2017 09.
Article in English | MEDLINE | ID: mdl-28572445

ABSTRACT

Polymicrobial sepsis in mice causes myocardial dysfunction after generation of the complement anaphylatoxin, complement component 5a (C5a). C5a interacts with its receptors on cardiomyocytes (CMs), resulting in redox imbalance and cardiac dysfunction that can be functionally measured and quantitated using Doppler echocardiography. In this report we have evaluated activation of MAPKs and Akt in CMs exposed to C5a in vitro and after cecal ligation and puncture (CLP) in vivo In both cases, C5a in vitro caused activation (phosphorylation) of MAPKs and Akt in CMs, which required availability of both C5a receptors. Using immunofluorescence technology, activation of MAPKs and Akt occurred in left ventricular (LV) CMs, requiring both C5a receptors, C5aR1 and -2. Use of a water-soluble p38 inhibitor curtailed activation in vivo of MAPKs and Akt in LV CMs as well as the appearance of cytokines and histones in plasma from CLP mice. When mouse macrophages were exposed in vitro to LPS, activation of MAPKs and Akt also occurred. The copresence of the p38 inhibitor blocked these activation responses. Finally, the presence of the p38 inhibitor in CLP mice reduced the development of cardiac dysfunction. These data suggest that polymicrobial sepsis causes cardiac dysfunction that appears to be linked to activation of MAPKs and Akt in heart.-Fattahi, F., Kalbitz, M., Malan, E. A., Abe, E., Jajou, L., Huber-Lang, M. S., Bosmann, M., Russell, M. W., Zetoune, F. S., Ward, P. A. Complement-induced activation of MAPKs and Akt during sepsis: role in cardiac dysfunction.


Subject(s)
Complement C5a/metabolism , Gene Expression Regulation/physiology , Heart Diseases/etiology , Mitogen-Activated Protein Kinase Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Sepsis/metabolism , Animals , Complement C5a/genetics , Heart Diseases/metabolism , Interleukins , Male , Mitogen-Activated Protein Kinase Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Rats , Rats, Sprague-Dawley , Receptor, Anaphylatoxin C5a/genetics , Receptor, Anaphylatoxin C5a/metabolism
9.
J Immunol ; 198(12): 4846-4854, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28490576

ABSTRACT

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.


Subject(s)
Complement Activation , Complement C5a/metabolism , Neutrophil Activation , Neutrophils/immunology , Sepsis/immunology , Sepsis/metabolism , Animals , Antacids/pharmacology , Calcium/metabolism , Calmodulin/metabolism , Complement C5a/immunology , Glucose/metabolism , Humans , Hydrogen-Ion Concentration , Lactates/metabolism , Lactoferrin , Mice , Neutrophils/chemistry , Neutrophils/drug effects , Neutrophils/metabolism , Peroxidase/metabolism , Protein Kinase C/immunology , Protein Kinase C/metabolism , Receptor, Anaphylatoxin C5a/metabolism , Signal Transduction
10.
Shock ; 48(1): 104-111, 2017 07.
Article in English | MEDLINE | ID: mdl-27926586

ABSTRACT

Blunt chest trauma induces severe local and systemic inflammatory alterations and an accumulation of apoptotic polymorphonuclear granulocytes (aPMN) in the lungs, frequently followed by bacterial infection. Alveolar macrophages (AM) represent one of the main actors for their clearance. However, little is known regarding regulatory and influencing factors of AM efferocytic and phagocytic activities. In this context, we investigated the influence of impaired gas exchange on AM activity.Male rats underwent blunt chest trauma or sham procedure and aPMN or Escherichia coli (E. coli) were instilled. Subsequently, the efferocytic and phagocytic activities were assessed by analyzing AM obtained from bronchoalveolar lavage fluids at three time points. To determine whether efferocytic and phagocytic activities of AM are affected by shifting gas concentrations, AM were subjected in vitro to hypoxic and hypercapnic conditions.Trauma significantly upregulated the capacity of AM to ingest E. coli starting 24 h after trauma, whereas the aPMN uptake rate remained virtually unchanged. In vitro, AM reacted to hypercapnic conditions by enhanced efferocytosis associated with increased release of anti-inflammatory cytokines. Additionally, phagocytosis and the pro-inflammatory reaction of AM after trauma appeared to be impaired. In contrast, hypoxic conditions displayed no regulatory effect on AM.In conclusion, blunt chest trauma enhances phagocytic activity of AM. On the other hand, hypercapnic conditions in the lungs may significantly contribute to the clearance of aPMN. The application of CO2 in clinical settings must be properly assessed, with the benefits of CO2 balanced against the detrimental effects of impaired bacterial clearance.


Subject(s)
Inflammation/immunology , Macrophages, Alveolar/immunology , Thoracic Injuries/immunology , Wounds, Nonpenetrating/immunology , Animals , Apoptosis/genetics , Apoptosis/physiology , Escherichia coli/pathogenicity , Granulocytes/immunology , Inflammation/microbiology , Male , Phagocytosis/genetics , Phagocytosis/physiology , Rats , Rats, Sprague-Dawley , Thoracic Injuries/microbiology , Wounds, Nonpenetrating/microbiology
11.
Shock ; 42(1): 3-10, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24667621

ABSTRACT

Acute lung injury (ALI) and its more severe form, acute respiratory distress syndrome (ARDS), can result from both direct and indirect pulmonary damage caused by trauma and shock. In the course of ALI/ARDS, mediators released from resident cells, such as alveolar macrophages, may act as chemoattractants for invading cells and stimulate local cells to build up a proinflammatory micromilieu. Depending on the trauma setting, the role of alveolar macrophages is differentially defined. This review focuses on alveolar macrophage function after blunt chest trauma, ischemia/reperfusion, hemorrhagic shock, and thermal burns.


Subject(s)
Acute Lung Injury/etiology , Macrophages, Alveolar/physiology , Thoracic Injuries/complications , Acute Lung Injury/pathology , Burns/complications , Burns/pathology , Humans , Inflammation Mediators/physiology , Lung/blood supply , Reperfusion Injury/complications , Respiratory Distress Syndrome/etiology , Respiratory Distress Syndrome/pathology , Shock, Hemorrhagic/complications , Thoracic Injuries/pathology , Wounds, Nonpenetrating/complications , Wounds, Nonpenetrating/pathology
12.
Immun Ageing ; 11(1): 20, 2014.
Article in English | MEDLINE | ID: mdl-25620994

ABSTRACT

BACKGROUND: Despite significant medical progress and improved treatment, surgical procedures of proximal femur fractures in older patients are still associated with a high postoperative complication and mortality rate. Recently, several authors investigated the phenomenon of immunoageing, indicating differences in the ageing immune system. The aim of the present multi-center prospective clinical trial was to analyze differences in the posttraumatic immune response of old patients compared to young patients. METHODS: Blood was collected from young patients (<50 y, n = 20) with long bone fractures (YF), old patients (>70 y, n = 21) with proximal femur fractures (OF) upon clinical admission and within 6 hours after surgery, and two healthy age matched control groups (YH & OH). Serum TRAIL- and cytokine concentrations were analyzed via cytometric bead array, Fas-Ligand and TNF-Receptor-I via ELISA. CD15(+) magnetic bead-isolated neutrophils (PMN) were TUNEL stained. RESULTS: IL-6 was significantly increased only in OF after trauma and surgery whereas YF patient exhibited a marked decrease of TNF after trauma. Interestingly, a significant increase of GM-CSF serum levels was observed in YF only, whereas OF exhibited a decrease of systemic IFN-γ concentrations after trauma and after surgery. The healthy controls, old and young, had more or less similar inflammation levels. Moreover, TRAIL serum levels were diminished in OF after trauma and even further after surgery whereas in YF this was only observed after the surgical procedure. Fas-L concentrations were reduced only in YF after surgery or trauma. PMN apoptosis was significantly reduced only in YF, indicating activation of the innate immune system. DISCUSSION: In summary, our data suggest that the posttraumatic immune response is differently regulated in old and young trauma patients. The operative procedure further impacts these differences after trauma. Whether the decreased activation of PMNs and phagocytes along with the observed dysregulation of the posttraumatic inflammatory response contributes to the high perioperative mortality rate of the elderly suffering from a proximal femoral fracture requires further investigation.

13.
PLoS One ; 8(5): e64454, 2013.
Article in English | MEDLINE | ID: mdl-23675538

ABSTRACT

Sympathoadrenergic progenitor cells (SAPs) of the peripheral nervous system (PNS) are important for normal development of the sympathetic PNS and for the genesis of neuroblastoma, the most common and often lethal extracranial solid tumor in childhood. However, it remains difficult to isolate sufficient numbers of SAPs for investigations. We therefore set out to improve generation of SAPs by using two complementary approaches, differentiation from murine embryonic stem cells (ESCs) and isolation from postnatal murine adrenal glands. We provide evidence that selecting for GD2 expression enriches for ESC-derived SAP-like cells and that proliferating SAP-like cells can be isolated from postnatal adrenal glands of mice. These advances may facilitate investigations about the development and malignant transformation of the sympathetic PNS.


Subject(s)
Adrenal Glands/cytology , Cell Differentiation , Embryonic Stem Cells/cytology , Neural Stem Cells/cytology , Sympathetic Nervous System/cytology , Adaptation, Physiological , Adrenal Glands/metabolism , Animals , Biomarkers/metabolism , CD57 Antigens/metabolism , Cell Culture Techniques , Cell Differentiation/physiology , Cell Line , Embryonic Stem Cells/metabolism , Gene Expression , Mice , Neural Stem Cells/metabolism , Neurons/cytology , Neurons/metabolism , Sympathetic Nervous System/metabolism
14.
Am J Physiol Lung Cell Mol Physiol ; 304(12): L863-72, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23564505

ABSTRACT

Zonulin is a protein involved in the regulation of tight junctions (TJ) in epithelial or endothelial cells. Zonulin is known to affect TJ in gut epithelial cells, but little is known about its influences in other organs. Prehaptoglobin2 has been identified as zonulin and is related to serine proteases (MASPs, C1qrs) that activate the complement system. The current study focused on the role of zonulin in development of acute lung injury (ALI) in C57BL/6 male mice following intrapulmonary deposition of IgG immune complexes. A zonulin antagonist (AT-1001) and a related peptide with permeability agonist activities (AT-1002) were employed and given intratracheally or intravenously. Also, zonulin was blocked in lung with a neutralizing antibody. In a dose-dependent manner, AT-1001 or zonulin neutralizing antibody attenuated the intensity of ALI (as quantitated by albumin leak, neutrophil accumulation, and proinflammatory cytokines). A similar pattern was found using the bacterial lipopolysaccharide model of ALI. Using confocal microscopy on sections of injured lungs, staining patterns for TJ proteins were discontinuous, reduced, and fragmented. As expected, the leak of blood products into the alveolar space confirmed the passage of 3 and 20 kDa dextran, and albumin. In contrast to AT-1001, application of the zonulin agonist AT-1002 intensified ALI. Zonulin both in vitro and in vivo induced generation of complement C3a and C5a. Collectively, these data suggest that zonulin facilitates development of ALI both by enhancing albumin leak and complement activation as well as increased buildup of neutrophils and cytokines during development of ALI.


Subject(s)
Acute Lung Injury/immunology , Cholera Toxin/genetics , Complement System Proteins/agonists , Protein Precursors/genetics , Acute Lung Injury/chemically induced , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Animals , Antigen-Antibody Complex/pharmacology , Cholera Toxin/agonists , Cholera Toxin/antagonists & inhibitors , Cholera Toxin/immunology , Complement Activation/drug effects , Complement System Proteins/immunology , Cytokines/biosynthesis , Cytokines/immunology , Gene Expression Regulation/drug effects , Haptoglobins , Immunoglobulin G/pharmacology , Lung/drug effects , Lung/immunology , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Neutrophils/drug effects , Neutrophils/immunology , Neutrophils/pathology , Oligopeptides/pharmacology , Peptides/pharmacology , Permeability/drug effects , Protein Precursors/agonists , Protein Precursors/antagonists & inhibitors , Protein Precursors/immunology , Signal Transduction/drug effects , Tight Junctions/drug effects , Tight Junctions/immunology , Tight Junctions/pathology , Trachea/drug effects , Trachea/immunology , Trachea/pathology
16.
J Trauma Acute Care Surg ; 74(3): 792-800, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23425737

ABSTRACT

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.


Subject(s)
Acute Lung Injury/etiology , Apoptosis , Bronchoalveolar Lavage Fluid/chemistry , Receptors, Tumor Necrosis Factor, Type I/metabolism , Sepsis/complications , Thoracic Injuries/complications , fas Receptor/metabolism , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Animals , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , In Situ Nick-End Labeling , Male , Mice , Mice, Inbred C57BL , Sepsis/metabolism , Sepsis/pathology , Signal Transduction , Thoracic Injuries/metabolism , Thoracic Injuries/pathology
17.
J Trauma Acute Care Surg ; 74(2): 489-98, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23354243

ABSTRACT

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.


Subject(s)
Apoptosis/physiology , Inflammation/etiology , Multiple Trauma/complications , Wounds and Injuries/complications , Animals , Blotting, Western , Brain/immunology , Brain/physiopathology , Bronchoalveolar Lavage Fluid/chemistry , Caspase 3/metabolism , Chemokines/analysis , Chemokines/blood , Cytokines/analysis , Cytokines/blood , Flow Cytometry , Hemoglobins/analysis , Inflammation/blood , Inflammation/immunology , Inflammation/physiopathology , Lung/chemistry , Lung/immunology , Lung/physiopathology , Male , Mice , Mice, Inbred C57BL , Multiple Trauma/blood , Multiple Trauma/immunology , Multiple Trauma/physiopathology , Peroxidase/metabolism , Wounds and Injuries/blood , Wounds and Injuries/immunology , Wounds and Injuries/physiopathology
18.
Shock ; 38(1): 98-106, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22552016

ABSTRACT

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.


Subject(s)
Acute Lung Injury/etiology , Neutrophil Activation/immunology , Neutrophils/immunology , Sepsis/complications , Thoracic Injuries/complications , Acute Lung Injury/immunology , Acute Lung Injury/pathology , Animals , Apoptosis/immunology , Bronchoalveolar Lavage Fluid/chemistry , Carbon Dioxide/blood , Caspases/metabolism , Chemokines/blood , Cytokines/blood , Disease Models, Animal , Lung/immunology , Male , Mice , Mice, Inbred C57BL , Oxygen/blood , Partial Pressure , Sepsis/immunology , Systemic Inflammatory Response Syndrome/etiology , Systemic Inflammatory Response Syndrome/immunology , Wounds, Nonpenetrating/complications , fas Receptor/blood
19.
PPAR Res ; 2012: 728461, 2012.
Article in English | MEDLINE | ID: mdl-22481914

ABSTRACT

Major trauma results in a strong inflammatory response in injured tissue. This posttraumatic hyperinflammation has been implied in the adverse events leading to a breakdown of host defense mechanisms and ultimately to delayed organ failure. Ligands to peroxisome proliferator-activated receptors (PPARs) have recently been identified as potent modulators of inflammation in various acute and chronic inflammatory conditions. The main mechanism of action mediated by ligand binding to PPARs is the inhibition of the nuclear transcription factor NF-κB, leading to downregulation of downstream gene transcription, such as for genes encoding proinflammatory cytokines. Pharmacological PPAR agonists exert strong anti-inflammatory properties in various animal models of tissue injury, including central nervous system trauma, ischemia/reperfusion injury, sepsis, and shock. In addition, PPAR agonists have been shown to induce wound healing process after tissue trauma. The present review was designed to provide an up-to-date overview on the current understanding of the role of PPARs in the pathophysiology of the inflammatory response after major trauma. Therapeutic options for using recombinant PPAR agonists as pharmacological agents in the management of posttraumatic inflammation will be discussed.

20.
Shock ; 37(2): 210-8, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22089186

ABSTRACT

Polymorphonuclear granulocytes (PMNs) have been attributed a primarily deleterious role in the pathogenesis of acute lung injury (ALI). However, evidence exists that PMNs might also act beneficially in certain types of ALI. In this regard, we investigated the role of activated neutrophils in the pathophysiology of lung contusion-induced ALI. We used the model of blunt chest trauma accompanied by PMN-depletion in male C3H/HeN mice. Animals received 25 µg/g body weight PMN-depleting antibody Gr-1 intravenously 48 h before trauma. Bronchoalveolar lavage (BAL) and lung tissue interleukin 6 (IL-6) were similarly elevated in PMN-depleted and control animals after trauma, whereas macrophage inflammatory protein 2 and monocyte chemoattractant protein 1 in BAL and lungs, IL-10 in BAL, and lung keratinocyte chemoattractant (KC) were even further increased in the absence of PMNs. Plasma IL-6 and KC were also increased in response to the insult and even further in the absence of PMNs. Chest trauma induced an enhanced release of IL-6, tumor necrosis factor α, macrophage inflammatory protein 2, monocyte chemoattractant protein 1, and IL-10 from isolated KU, which was blunted in the absence of PMNs. In the presence of PMNs, BAL protein was further increased at 30 h when compared with the 3-h time point, which was not the case in the absence of PMNs. Taken together, in response to lung trauma, activated neutrophils control inflammation including mediator release from distant immune cells but simultaneously mediate pulmonary tissue damage. Thus, keeping in mind potential inflammatory adverse effects, modulation of neutrophil activation or trafficking might be a reasonable therapeutic approach in chest trauma-induced lung injury.


Subject(s)
Granulocytes/cytology , Kupffer Cells/cytology , Lung Injury/metabolism , Neutrophils/cytology , Spleen/cytology , Animals , Body Weight , Bronchoalveolar Lavage , Keratinocytes/cytology , Male , Mice , Mice, Inbred C3H , Peroxidase/metabolism , Time Factors , Wounds, Nonpenetrating/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...