Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Scand J Immunol ; 91(2): e12837, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31622512

RESUMO

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.


Assuntos
Monitorização Imunológica/métodos , Traumatismo Múltiplo/diagnóstico , Adulto , Células Cultivadas , Progressão da Doença , Feminino , Humanos , Lipopolissacarídeos/imunologia , Masculino , Pessoa de Meia-Idade , Projetos Piloto
2.
J Immunol ; 198(12): 4846-4854, 2017 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-28490576

RESUMO

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.


Assuntos
Ativação do Complemento , Complemento C5a/metabolismo , Ativação de Neutrófilo , Neutrófilos/imunologia , Sepse/imunologia , Sepse/metabolismo , Animais , Antiácidos/farmacologia , Cálcio/metabolismo , Calmodulina/metabolismo , Complemento C5a/imunologia , Glucose/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Lactatos/metabolismo , Lactoferrina , Camundongos , Neutrófilos/química , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Peroxidase/metabolismo , Proteína Quinase C/imunologia , Proteína Quinase C/metabolismo , Receptor da Anafilatoxina C5a/metabolismo , Transdução de Sinais
3.
FASEB J ; 31(9): 4129-4139, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28572445

RESUMO

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.


Assuntos
Complemento C5a/metabolismo , Regulação da Expressão Gênica/fisiologia , Cardiopatias/etiologia , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Sepse/metabolismo , Animais , Complemento C5a/genética , Cardiopatias/metabolismo , Interleucinas , Masculino , Quinases de Proteína Quinase Ativadas por Mitógeno/genética , Proteínas Proto-Oncogênicas c-akt/genética , Ratos , Ratos Sprague-Dawley , Receptor da Anafilatoxina C5a/genética , Receptor da Anafilatoxina C5a/metabolismo
4.
Mediators Inflamm ; 2018: 2052356, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30002598

RESUMO

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.


Assuntos
Complemento C5a/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Choque Hemorrágico/metabolismo , Animais , Células Cultivadas , Relação Dose-Resposta a Droga , Eletrofisiologia , Citometria de Fluxo , Humanos , Concentração de Íons de Hidrogênio , Masculino , Suínos
5.
Immun Ageing ; 11(1): 20, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25620994

RESUMO

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.

6.
Am J Physiol Lung Cell Mol Physiol ; 304(12): L863-72, 2013 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-23564505

RESUMO

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.


Assuntos
Lesão Pulmonar Aguda/imunologia , Toxina da Cólera/genética , Proteínas do Sistema Complemento/agonistas , Precursores de Proteínas/genética , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/metabolismo , Lesão Pulmonar Aguda/patologia , Animais , Complexo Antígeno-Anticorpo/farmacologia , Toxina da Cólera/agonistas , Toxina da Cólera/antagonistas & inibidores , Toxina da Cólera/imunologia , Ativação do Complemento/efeitos dos fármacos , Proteínas do Sistema Complemento/imunologia , Citocinas/biossíntese , Citocinas/imunologia , Regulação da Expressão Gênica/efeitos dos fármacos , Haptoglobinas , Imunoglobulina G/farmacologia , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Pulmão/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/efeitos dos fármacos , Neutrófilos/imunologia , Neutrófilos/patologia , Oligopeptídeos/farmacologia , Peptídeos/farmacologia , Permeabilidade/efeitos dos fármacos , Precursores de Proteínas/agonistas , Precursores de Proteínas/antagonistas & inibidores , Precursores de Proteínas/imunologia , Transdução de Sinais/efeitos dos fármacos , Junções Íntimas/efeitos dos fármacos , Junções Íntimas/imunologia , Junções Íntimas/patologia , Traqueia/efeitos dos fármacos , Traqueia/imunologia , Traqueia/patologia
7.
Nature ; 449(7163): 721-5, 2007 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-17914358

RESUMO

It is becoming increasingly clear that the autonomic nervous system and the immune system demonstrate cross-talk during inflammation by means of sympathetic and parasympathetic pathways. We investigated whether phagocytes are capable of de novo production of catecholamines, suggesting an autocrine/paracrine self-regulatory mechanism by catecholamines during inflammation, as has been described for lymphocytes. Here we show that exposure of phagocytes to lipopolysaccharide led to a release of catecholamines and an induction of catecholamine-generating and degrading enzymes, indicating the presence of the complete intracellular machinery for the generation, release and inactivation of catecholamines. To assess the importance of these findings in vivo, we chose two models of acute lung injury. Blockade of alpha2-adrenoreceptors or catecholamine-generating enzymes greatly suppressed lung inflammation, whereas the opposite was the case either for an alpha2-adrenoreceptor agonist or for inhibition of catecholamine-degrading enzymes. We were able to exclude T cells or sympathetic nerve endings as sources of the injury-modulating catecholamines. Our studies identify phagocytes as a new source of catecholamines, which enhance the inflammatory response.


Assuntos
Catecolaminas/metabolismo , Pneumopatias/metabolismo , Pneumopatias/patologia , Fagócitos/metabolismo , Doença Aguda , Antagonistas de Receptores Adrenérgicos alfa 2 , Animais , Inflamação/metabolismo , Inflamação/patologia , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Fagócitos/efeitos dos fármacos , Fagócitos/enzimologia , Ratos , Ratos Long-Evans , Receptores Adrenérgicos alfa 2/metabolismo , Linfócitos T/metabolismo
8.
J Cell Biol ; 222(2)2023 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-36459066

RESUMO

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.


Assuntos
Apoptose , Antígeno CD47 , Macrófagos , Fenótipo Secretor Associado à Senescência , Animais , Humanos , Camundongos , Aminoaciltransferases/metabolismo , Antígeno CD24/metabolismo , Antígeno CD47/genética , Antígeno CD47/metabolismo , Macrófagos/citologia , Regulação para Cima
9.
PLoS Pathog ; 5(6): e1000464, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19503602

RESUMO

Pathogen-pattern-recognition by Toll-like receptors (TLRs) and pathogen clearance after immune complex formation via engagement with Fc receptors (FcRs) represent central mechanisms that trigger the immune and inflammatory responses. In the present study, a linkage between TLR4 and FcgammaR was evaluated in vitro and in vivo. Most strikingly, in vitro activation of phagocytes by IgG immune complexes (IgGIC) resulted in an association of TLR4 with FcgammaRIII (CD16) based on co-immunoprecipitation analyses. Neutrophils and macrophages from TLR4 mutant (mut) mice were unresponsive to either lipopolysaccharide (LPS) or IgGIC in vitro, as determined by cytokine production. This phenomenon was accompanied by the inability to phosphorylate tyrosine residues within immunoreceptor tyrosine-based activation motifs (ITAMs) of the FcRgamma-subunit. To transfer these findings in vivo, two different models of acute lung injury (ALI) induced by intratracheal administration of either LPS or IgGIC were employed. As expected, LPS-induced ALI was abolished in TLR4 mut and TLR4(-/-) mice. Unexpectedly, TLR4 mut and TLR4(-/-) mice were also resistant to development of ALI following IgGIC deposition in the lungs. In conclusion, our findings suggest that TLR4 and FcgammaRIII pathways are structurally and functionally connected at the receptor level and that TLR4 is indispensable for FcgammaRIII signaling via FcRgamma-subunit activation.


Assuntos
Lesão Pulmonar Aguda/imunologia , Receptor Cross-Talk , Receptores de IgG/metabolismo , Transdução de Sinais , Receptor 4 Toll-Like/metabolismo , Lesão Pulmonar Aguda/metabolismo , Análise de Variância , Animais , Complexo Antígeno-Anticorpo/imunologia , Células Cultivadas , Citocinas/metabolismo , Imunoglobulina G/imunologia , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Lipopolissacarídeos/imunologia , Pulmão/imunologia , Pulmão/metabolismo , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/metabolismo , Masculino , Camundongos , Camundongos Knockout , Fosforilação , Receptor da Anafilatoxina C5a , Receptores de Complemento/genética , Receptores de Complemento/metabolismo , Receptores de IgG/genética , Receptor 4 Toll-Like/genética
10.
J Biomed Sci ; 18: 90, 2011 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-22129197

RESUMO

Trauma represents the leading cause of death among young people in industrialized countries. Recent clinical and experimental studies have brought increasing evidence for activation of the innate immune system in contributing to the pathogenesis of trauma-induced sequelae and adverse outcome. As the "first line of defense", the complement system represents a potent effector arm of innate immunity, and has been implicated in mediating the early posttraumatic inflammatory response. Despite its generic beneficial functions, including pathogen elimination and immediate response to danger signals, complement activation may exert detrimental effects after trauma, in terms of mounting an "innocent bystander" attack on host tissue. Posttraumatic ischemia/reperfusion injuries represent the classic entity of complement-mediated tissue damage, adding to the "antigenic load" by exacerbation of local and systemic inflammation and release of toxic mediators. These pathophysiological sequelae have been shown to sustain the systemic inflammatory response syndrome after major trauma, and can ultimately contribute to remote organ injury and death. Numerous experimental models have been designed in recent years with the aim of mimicking the inflammatory reaction after trauma and to allow the testing of new pharmacological approaches, including the emergent concept of site-targeted complement inhibition. The present review provides an overview on the current understanding of the cellular and molecular mechanisms of complement activation after major trauma, with an emphasis of emerging therapeutic concepts which may provide the rationale for a "bench-to-bedside" approach in the design of future pharmacological strategies.


Assuntos
Proteínas do Sistema Complemento/fisiologia , Inflamação/imunologia , Ferimentos e Lesões/imunologia , Animais , Ativação do Complemento/imunologia , Proteínas do Sistema Complemento/imunologia , Humanos , Imunidade Inata
11.
Langenbecks Arch Surg ; 396(2): 251-9, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20393744

RESUMO

PURPOSE: The purpose of this study was to characterize the local pulmonary inflammatory environment and to elucidate alterations of alveolar macrophage (AMØ) functions after blunt chest trauma. METHODS: Wistar rats were subjected to blunt chest trauma. AMØ were isolated, stimulated, and cultured. Bronchoalveolar lavage (BAL) was collected. Cytokines/chemokines were quantified in the BAL and in AMØ supernatants via ELISA. AMØ phagocytic and chemotactic activity and respiratory burst capacity were assessed. RESULTS: Following chest trauma, a significant increase of IL-1ß (at 6 and 24 h) and IL-6 (at 24 h) in BAL was observed, whereas IL-10 and TNF-α concentrations were not altered. MIP-2 and CINC were substantially increased as early as 6 h and PGE2 early at 10 min, whereas BAL MCP-1 was not elevated until 24 h after trauma. MIP-2 release by AMØ isolated form trauma animals was markedly increased as early as 10 min after injury. IL-1ß and IL-10 exhibited a late increase at 24 h. AMØ TNF-α release was increased at 6 h. At 6 or 24 h, AMØ from trauma animals incorporated significantly more opsonized latex beads than their sham controls, and their chemotactic activity was substantially enhanced at 24 h. AMØ oxidative burst capacity remained largely unchanged. CONCLUSIONS: Already very early after chest trauma, inflammatory mediators are present in the intraalveolar compartment. Additionally, AMØ are primed to release cytokines and chemokines. Blunt chest trauma also changes the phagocytic and chemotactic activity of AMØ. These functional changes of AMØ might enable them to better ward off potential pathogens in the course after trauma.


Assuntos
Citocinas/imunologia , Macrófagos Alveolares/imunologia , Traumatismos Torácicos/imunologia , Animais , Quimiotaxia , Modelos Animais de Doenças , Macrófagos Alveolares/metabolismo , Masculino , Fagocitose , Ratos , Ratos Wistar , Explosão Respiratória , Ferimentos não Penetrantes/imunologia
12.
Br J Pharmacol ; 178(14): 2863-2879, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-32880897

RESUMO

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.


Assuntos
Proteínas do Sistema Complemento , Sistema Imunitário , Humanos , Hipóxia
13.
Crit Care ; 14(3): 165, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20565858

RESUMO

The exact cellular and molecular mechanisms of sepsis-induced encephalopathy remain elusive. The breakdown of the blood-brain barrier (BBB) is considered a focal point in the development of sepsis-induced brain damage. Contributing factors for the compromise of the BBB include cytokines and chemokines, activation of the complement cascade, phagocyte-derived toxic mediators, and bacterial products. To date, we are far from fully understanding the neuropathology that develops as a secondary remote organ injury as a consequence of sepsis. However, recent studies suggest that bacterial proteins may readily cross the functional BBB and trigger an inflammatory response in the subarachnoid space, in absence of a bacterial invasion. A better understanding of the pathophysiological events leading to septic encephalopathy appears crucial to advance the clinical care for this vulnerable patient population.


Assuntos
Barreira Hematoencefálica/microbiologia , Infecções Bacterianas do Sistema Nervoso Central/etiologia , Hipóxia-Isquemia Encefálica/fisiopatologia , Sepse/complicações , Biomarcadores , Infecções Bacterianas do Sistema Nervoso Central/imunologia , Infecções Bacterianas do Sistema Nervoso Central/fisiopatologia , Cuidados Críticos , Humanos , Microcirculação , Sepse/imunologia , Sepse/fisiopatologia
14.
Front Immunol ; 11: 1789, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32983087

RESUMO

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.


Assuntos
Micropartículas Derivadas de Células/imunologia , Complemento C5a/metabolismo , Imunidade Inata , Mediadores da Inflamação/metabolismo , Traumatismo Múltiplo/imunologia , Neutrófilos/imunologia , Fator 6 de Ribosilação do ADP , Fatores de Ribosilação do ADP/metabolismo , Adulto , Estudos de Casos e Controles , Micropartículas Derivadas de Células/metabolismo , Feminino , Humanos , Escala de Gravidade do Ferimento , Interleucina-6/metabolismo , Cinética , Masculino , Pessoa de Meia-Idade , Traumatismo Múltiplo/sangue , Traumatismo Múltiplo/diagnóstico , NADP/metabolismo , Neutrófilos/metabolismo , Peroxidase/metabolismo , Estudos Prospectivos , Espécies Reativas de Oxigênio/metabolismo , Receptor da Anafilatoxina C5a/metabolismo , Transdução de Sinais , Adulto Jovem
15.
FASEB J ; 22(10): 3483-90, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18587006

RESUMO

Activation of the complement system is a key event in the pathogenesis of sepsis. Nevertheless, the exact mechanisms remain inadequately understood. In the current study, we examined the role of complement C3 and C5 in sepsis in wild-type and C3- or C5-deficient mice induced by cecal ligation and puncture. When compared to wild-type mice, C5(-/-) showed identical survival, and C3(-/-) presented significantly reduced survival. Interestingly, this was associated with significant decreases in plasma levels of proinflammatory mediators. Moreover, although septic C3(-/-) animals displayed a 10-fold increase of blood-borne bacteria, C5(-/-) animals exhibited a 400-fold increase in bacteremia when compared to wild-type mice. These effects were linked to the inability of C5(-/-) mice to assemble the terminal membrane attack complex (MAC), as determined by complement hemolytic activity (CH-50). Surprisingly, although negative control C3(-/-) mice failed to generate the MAC, significant increases of MAC formation was found in septic C3(-/-) mice. In conclusion, our data corroborate that hemolytic complement activity is essential for control of bacteremia in septic mice. Thus, during sepsis, blockade of C5a or its receptors (rather than C5) seems a more promising strategy, because C5a-blockade still allows for MAC formation while the adverse effects of C5a are prevented.


Assuntos
Bacteriemia/imunologia , Complemento C3/fisiologia , Complemento C5a/fisiologia , Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Animais , Bacteriemia/genética , Quimiocinas/sangue , Complemento C3/genética , Complemento C5a/genética , Ensaio de Atividade Hemolítica de Complemento , Citocinas/sangue , Modelos Animais de Doenças , Camundongos , Camundongos Knockout
16.
FASEB J ; 22(7): 2198-205, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18299333

RESUMO

IL-17A is a proinflammatory cytokine produced by a variety of cells. In the current study, we examined the role of IL-17A in sepsis induced in mice by cecal ligation and puncture (CLP). IL-17A levels, which rose time-dependently in plasma after CLP, were not affected in the absence of alphabeta T cells or neutrophils. In sharp contrast, gammadelta T cell-knockout or gammadelta T cell-depleted mice displayed baseline IL-17A plasma levels after CLP. Neutralization of IL-17A by two different antibodies improved sepsis (survival from approximately 10% to nearly 60%). Unexpectedly, antibody treatment was protective, even when administration of anti-IL-17A was delayed for up to 12 h after CLP. These protective effects of IL-17A blockade were associated with substantially reduced levels of bacteremia together with significant reductions of systemic proinflammatory cytokines and chemokines in plasma. In vitro incubation of mouse peritoneal macrophages with lipopolysaccharide (LPS) in the copresence of IL-17A substantially increased the production of TNF-alpha, IL-1beta, and IL-6 by these cells. These data suggest that, during experimental sepsis, gammadelta T cell-derived IL-17A promotes high levels of proinflammatory mediators and bacteremia, resulting in enhanced lethality. IL-17A may be a potential therapeutic target in sepsis.


Assuntos
Interleucina-17/toxicidade , Sepse/fisiopatologia , Animais , Bacteriemia , Ceco/patologia , Quimiocinas/sangue , Citocinas/sangue , Modelos Animais de Doenças , Inflamação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Punções , Sepse/etiologia , Linfócitos T/patologia
17.
Shock ; 29(1): 25-31, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17621257

RESUMO

The inflammatory response after severe blunt chest trauma often leads to acute lung injury and acute respiratory distress syndrome which are associated with high mortality rates. Whereas the role of innate immunity in acute lung injury has been broadly investigated, the immune response after blunt chest trauma is still poorly understood. Therefore, the role of complement and neutrophils was determined in bilateral lung injury induced by a single blast wave. The following time-points were investigated posttrauma: sham, 1, 6, 12, and 24 h. There was a time-dependent systemic activation of complement as determined by CH-50 and presence of C5a-dependent chemotactic plasma activity. Moreover, factor H, a complement regulatory protein, was increased systemically and locally after injury. Anti-C5a treatment immediately after trauma ameliorated these peaks. After an initial systemic leukopenic phase, a marked leukocytosis occurred. The latter was normalized by C5a blockade. In parallel, white blood cell count in bronchioalveolar lavage fluids was increased as a function of time and was significantly decreased by anti-C5a treatment. Trauma-induced lung injury was also associated with dramatic changes in neutrophil function, namely early enhanced chemotaxis and phagocytosis, followed by prolonged functional defects-all of which were ameliorated by anti-C5a treatment. Furthermore, blockade of C5a ameliorated the buildup of the proinflammatory cytokine TNF-alpha, diminished the increase of cytokine-induced neutrophil chemoattractant 1, and altered the levels of the anti-inflammatory cytokine IL-10. These data suggest that blunt chest trauma leads to systemic activation of complement and robust C5a generation, which causes perturbations in defensive functions of neutrophils. Thus, C5a might represent a potential target for therapeutic immunomodulation to prevent immune dysfunctions post-trauma and thereby, perhaps, the progression to acute respiratory distress syndrome.


Assuntos
Complemento C5a/imunologia , Imunidade Inata , Traumatismos Torácicos/imunologia , Ferimentos não Penetrantes/imunologia , Animais , Líquido da Lavagem Broncoalveolar/citologia , Líquido da Lavagem Broncoalveolar/imunologia , Quimiocina CXCL1/metabolismo , Quimiotaxia de Leucócito , Ativação do Complemento , Complemento C5a/antagonistas & inibidores , Fator H do Complemento/metabolismo , Modelos Animais de Doenças , Humanos , Interleucina-10/metabolismo , Neutrófilos/imunologia , Fagocitose , Ratos , Ratos Wistar , Explosão Respiratória , Fator de Necrose Tumoral alfa/metabolismo
20.
PLoS One ; 12(10): e0187327, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29084268

RESUMO

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.


Assuntos
Coração/fisiopatologia , Traumatismo Múltiplo/patologia , Choque Hemorrágico/patologia , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA