Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Semin Immunol ; 28(3): 278-84, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27157701

RESUMO

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


Assuntos
Inativadores do Complemento/uso terapêutico , Imunoterapia/métodos , Inflamação/terapia , Choque Hemorrágico/terapia , Ferimentos e Lesões/terapia , Animais , Modelos Animais de Doenças , Humanos , Imunidade Inata , Inflamação/imunologia , Camundongos , Camundongos Knockout , Moléculas com Motivos Associados a Patógenos/imunologia , Choque Hemorrágico/imunologia , Ferimentos e Lesões/imunologia
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.
Mediators Inflamm ; 2015: 463950, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26556956

RESUMO

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


Assuntos
Moléculas de Adesão Celular/sangue , Traumatismo Múltiplo/sangue , Receptores de Superfície Celular/sangue , APACHE , Animais , Líquido da Lavagem Broncoalveolar/química , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Índice de Gravidade de Doença
4.
Semin Immunopathol ; 40(1): 87-102, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28900700

RESUMO

Activation and regulation of the cascade systems of the blood (the complement system, the coagulation/contact activation/kallikrein system, and the fibrinolytic system) occurs via activation of zymogen molecules to specific active proteolytic enzymes. Despite the fact that the generated proteases are all present together in the blood, under physiological conditions, the activity of the generated proteases is controlled by endogenous protease inhibitors. Consequently, there is remarkable little crosstalk between the different systems in the fluid phase. This concept review article aims at identifying and describing conditions where the strict system-related control is circumvented. These include clinical settings where massive amounts of proteolytic enzymes are released from tissues, e.g., during pancreatitis or post-traumatic tissue damage, resulting in consumption of the natural substrates of the specific proteases and the available protease inhibitor. Another example of cascade system dysregulation is disseminated intravascular coagulation, with canonical activation of all cascade systems of the blood, also leading to specific substrate and protease inhibitor elimination. The present review explains basic concepts in protease biochemistry of importance to understand clinical conditions with extensive protease activation.


Assuntos
Ativação do Complemento/imunologia , Proteínas do Sistema Complemento/imunologia , Proteínas do Sistema Complemento/metabolismo , Suscetibilidade a Doenças/imunologia , Animais , Convertases de Complemento C3-C5 da Via Clássica/imunologia , Convertases de Complemento C3-C5 da Via Clássica/metabolismo , Ativação Enzimática , Humanos , Cinética , Terapia de Alvo Molecular , Peptídeo Hidrolases/metabolismo , Inibidores de Proteases/sangue , Inibidores de Proteases/metabolismo , Ligação Proteica , Proteólise , Transdução de Sinais , Especificidade por Substrato
5.
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
6.
Immunobiology ; 221(10): 1073-9, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27371975

RESUMO

The humoral serine proteases of the complement system and the coagulation system play central roles during the events of an inflammatory response. While the complement system confers immunoprotective and -regulatory functions, the coagulation cascade is responsible to ensure hemostatic maintenance. Although these two systems individually unfold during inflammation, several studies have reported on the "crosstalk" between components of the complement and the coagulation system in the fluid phase. However, both cascades are usually initiated on or in close proximity to foreign or activated surfaces, and there is increasing evidence for interacting complement and coagulation proteins on various superficial areas on endothelium, circulating entities like platelets, leukocytes, microparticles and pathogens, and even on artificial surfaces. This review aims at summarizing these interactions to complete the picture.


Assuntos
Coagulação Sanguínea , Proteínas do Sistema Complemento/fisiologia , Transdução de Sinais , Animais , Células Sanguíneas/imunologia , Células Sanguíneas/metabolismo , Fatores de Coagulação Sanguínea/metabolismo , Humanos , Ligação Proteica
7.
Stem Cells Int ; 2016: 6289825, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27340408

RESUMO

Mesenchymal stem cells (MSCs) are multipotent cells that are considered indispensable in regeneration processes after tissue trauma. MSCs are recruited to damaged areas via several chemoattractant pathways where they function as "actors" in the healing process by the secretion of manifold pro- and anti-inflammatory, antimicrobial, pro- and anticoagulatory, and trophic/angiogenic factors, but also by proliferation and differentiation into the required cells. On the other hand, MSCs represent "targets" during the pathophysiological conditions after severe trauma, when excessively generated inflammatory mediators, complement activation factors, and damage- and pathogen-associated molecular patterns challenge MSCs and alter their functionality. This in turn leads to complement opsonization, lysis, clearance by macrophages, and reduced migratory and regenerative abilities which culminate in impaired tissue repair. We summarize relevant cellular and signaling mechanisms and provide an up-to-date overview about promising future therapeutic MSC strategies in the context of severe tissue trauma.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA