Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Cell Mol Med ; 19(11): 2549-63, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26337158

RESUMO

Acute respiratory distress syndrome (ARDS) induced by severe sepsis can trigger persistent inflammation and fibrosis. We have shown that experimental sepsis in baboons recapitulates ARDS progression in humans, including chronic inflammation and long-lasting fibrosis in the lung. Complement activation products may contribute to the fibroproliferative response, suggesting that complement inhibitors are potential therapeutic agents. We have been suggested that treatment of septic baboons with compstatin, a C3 convertase inhibitor protects against ARDS-induced fibroproliferation. Baboons challenged with 10(9) cfu/kg (LD50) live E. coli by intravenous infusion were treated or not with compstatin at the time of challenge or 5 hrs thereafter. Changes in the fibroproliferative response at 24 hrs post-challenge were analysed at both transcript and protein levels. Gene expression analysis showed that sepsis induced fibrotic responses in the lung as early as 24 hrs post-bacterial challenge. Immunochemical and biochemical analysis revealed enhanced collagen synthesis, induction of profibrotic factors and increased cell recruitment and proliferation. Specific inhibition of complement with compstatin down-regulated sepsis-induced fibrosis genes, including transforming growth factor-beta (TGF-ß), connective tissue growth factor (CTGF), tissue inhibitor of metalloproteinase 1 (TIMP1), various collagens and chemokines responsible for fibrocyte recruitment (e.g. chemokine (C-C motif) ligand 2 (CCL2) and 12 (CCL12)). Compstatin decreased the accumulation of myofibroblasts and proliferating cells, reduced the production of fibrosis mediators (TGF-ß, phospho-Smad-2 and CTGF) and inhibited collagen deposition. Our data demonstrate that complement inhibition effectively attenuates collagen deposition and fibrotic responses in the lung after severe sepsis. Inhibiting complement could prove an attractive strategy for preventing sepsis-induced fibrosis of the lung.


Assuntos
Bacteriemia/tratamento farmacológico , Ativação do Complemento/efeitos dos fármacos , Inativadores do Complemento/uso terapêutico , Infecções por Escherichia coli/tratamento farmacológico , Pulmão/patologia , Peptídeos Cíclicos/uso terapêutico , Animais , Bacteriemia/imunologia , Bacteriemia/patologia , Infecções por Escherichia coli/imunologia , Infecções por Escherichia coli/fisiopatologia , Fibrose , Regulação da Expressão Gênica/efeitos dos fármacos , Síndrome do Desconforto Respiratório/tratamento farmacológico , Síndrome do Desconforto Respiratório/imunologia , Síndrome do Desconforto Respiratório/patologia
2.
Am J Respir Cell Mol Biol ; 50(2): 439-50, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24066737

RESUMO

Sepsis-induced inflammation of the lung leads to acute respiratory distress syndrome (ARDS), which may trigger persistent fibrosis. The pathology of ARDS is complex and poorly understood, and the therapeutic approaches are limited. We used a baboon model of Escherichia coli sepsis that mimics the complexity of human disease to study the pathophysiology of ARDS. We performed extensive biochemical, histological, and functional analyses to characterize the disease progression and the long-term effects of sepsis on the lung structure and function. Similar to humans, sepsis-induced ARDS in baboons displays an early inflammatory exudative phase, with extensive necrosis. This is followed by a regenerative phase dominated by proliferation of type 2 epithelial cells, expression of epithelial-to-mesenchymal transition markers, myofibroblast migration and proliferation, and collagen synthesis. Baboons that survived sepsis showed persistent inflammation and collagen deposition 6-27 months after the acute episodes. Long-term survivors had almost double the amount of collagen in the lung as compared with age-matched control animals. Immunostaining for procollagens showed persistent active collagen synthesis within the fibroblastic foci and interalveolar septa. Fibroblasts expressed markers of transforming growth factor-ß and platelet-derived growth factor signaling, suggesting their potential role as mediators of myofibroblast migration and proliferation, and collagen deposition. In parallel, up-regulation of the inhibitors of extracellular proteases supports a deregulated matrix remodeling that may contribute to fibrosis. The primate model of sepsis-induced ARDS mimics the disease progression in humans, including chronic inflammation and long-lasting fibrosis. This model helps our understanding of the pathophysiology of fibrosis and the testing of new therapies.


Assuntos
Lesão Pulmonar Aguda/metabolismo , Escherichia coli , Síndrome do Desconforto Respiratório/metabolismo , Sepse/metabolismo , Lesão Pulmonar Aguda/fisiopatologia , Animais , Colágeno/metabolismo , Modelos Animais de Doenças , Fibrose/metabolismo , Humanos , Inflamação/metabolismo , Inflamação/patologia , Pulmão/metabolismo , Pulmão/patologia , Papio , Síndrome do Desconforto Respiratório/patologia , Síndrome do Desconforto Respiratório/fisiopatologia , Sepse/patologia , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/metabolismo
3.
Blood ; 116(6): 1002-10, 2010 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-20466856

RESUMO

Severe sepsis leads to massive activation of coagulation and complement cascades that could contribute to multiple organ failure and death. To investigate the role of the complement and its crosstalk with the hemostatic system in the pathophysiology and therapeutics of sepsis, we have used a potent inhibitor (compstatin) administered early or late after Escherichia coli challenge in a baboon model of sepsis-induced multiple organ failure. Compstatin infusion inhibited sepsis-induced blood and tissue biomarkers of complement activation, reduced leucopenia and thrombocytopenia, and lowered the accumulation of macrophages and platelets in organs. Compstatin decreased the coagulopathic response by down-regulating tissue factor and PAI-1, diminished global blood coagulation markers (fibrinogen, fibrin-degradation products, APTT), and preserved the endothelial anticoagulant properties. Compstatin treatment also improved cardiac function and the biochemical markers of kidney and liver damage. Histologic analysis of vital organs collected from animals euthanized after 24 hours showed decreased microvascular thrombosis, improved vascular barrier function, and less leukocyte infiltration and cell death, all consistent with attenuated organ injury. We conclude that complement-coagulation interplay contributes to the progression of severe sepsis and blocking the harmful effects of complement activation products, especially during the organ failure stage of severe sepsis is a potentially important therapeutic strategy.


Assuntos
Coagulação Sanguínea/efeitos dos fármacos , Proteínas Inativadoras do Complemento/farmacologia , Infecções por Escherichia coli , Insuficiência de Múltiplos Órgãos/prevenção & controle , Peptídeos Cíclicos/farmacologia , Sepse , Animais , Biomarcadores/sangue , Coagulação Sanguínea/imunologia , Pressão Sanguínea/efeitos dos fármacos , Ativação do Complemento/efeitos dos fármacos , Proteínas Inativadoras do Complemento/metabolismo , Citocinas/sangue , Modelos Animais de Doenças , Infecções por Escherichia coli/sangue , Infecções por Escherichia coli/tratamento farmacológico , Infecções por Escherichia coli/imunologia , Insuficiência de Múltiplos Órgãos/sangue , Insuficiência de Múltiplos Órgãos/imunologia , Papio , Sepse/sangue , Sepse/tratamento farmacológico , Sepse/imunologia
4.
Am J Pathol ; 171(3): 1066-77, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17640967

RESUMO

Increased tissue factor (TF)-dependent procoagulant activity in sepsis may be partly due to decreased expression or function of tissue factor pathway inhibitor (TFPI). To test this hypothesis, baboons were infused with live Escherichia coli and sacrificed after 2, 8, or 24 hours. Confocal and electron microscopy revealed increased leukocyte infiltration and fibrin deposition in the intravascular and interstitial compartments. Large amounts of TF were detected by immunostaining in leukocytes and platelet-rich microthrombi. TF induction was documented by quantitative reverse transcriptase-polymerase chain reaction, enzyme-linked immunosorbent assay, and coagulation assays. Lung-associated TFPI antigen and mRNA decreased during sepsis, and TFPI activity diminished abruptly at 2 hours. Blocking antibodies against TFPI increased fibrin deposition in septic baboon lungs, suggesting that TF-dependent coagulation might be aggravated by reduced endothelial TFPI. Decreased TFPI activity coincided with the release of tissue plasminogen activator and the peak of plasmin generation, suggesting that TFPI could undergo proteolytic inactivation by plasmin. Enhanced plasmin produced in septic baboons by infusion of blocking antibodies against plasminogen activator inhibitor-1 led to decreased lung-associated TFPI and unforeseen massive fibrin deposition. We conclude that activation of TF-driven coagulation not adequately countered by TFPI may underlie the widespread thrombotic complications of sepsis.


Assuntos
Anticoagulantes/metabolismo , Coagulação Sanguínea , Lipoproteínas/metabolismo , Pulmão/metabolismo , Pulmão/patologia , Papio cynocephalus , Sepse , Animais , Anticorpos/metabolismo , Escherichia coli/imunologia , Fibrinolisina/metabolismo , Humanos , Pulmão/citologia , Pulmão/microbiologia , Macrófagos/citologia , Macrófagos/metabolismo , Neutrófilos/citologia , Neutrófilos/metabolismo , Inibidor 1 de Ativador de Plasminogênio/metabolismo
5.
Blood ; 110(9): 3168-75, 2007 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-17644733

RESUMO

Thrombin activatable fibrinolysis inhibitor (TAFI), when activated, forms a basic carboxypeptidase that can inhibit fibrinolysis. Potential physiologic activators include both thrombin and plasmin. In vitro, thrombomodulin and glycosaminoglycans increase the catalytic efficiency of TAFI activation by thrombin and plasmin, respectively. The most relevant (patho-) physiologic activator of TAFI has not been disclosed. Our purpose was to identify the physiologic activator of TAFI in vivo. Activation of protein C (a thrombin-thrombomodulin-dependent reaction), prothrombin, and plasminogen occurs during sepsis. Thus, a baboon model of Escherichia coli-induced sepsis, where multiple potential activators of TAFI are elaborated, was used to study TAFI activation. A monoclonal antibody (mAbTAFI/TM#16) specifically inhibiting thrombin-thrombomodulin-dependent activation of TAFI was used to assess the contribution of thrombin-thrombomodulin in TAFI activation in vivo. Coinfusion of mAbTAFI/TM#16 with a lethal dose of E coli prevented the complete consumption of TAFI observed without mAbTAFI/TM#16. The rate of fibrin degradation products formation is enhanced in septic baboons treated with the mAbTAFI/TM#16; therefore, TAFI activation appears to play a key role in the extent of fibrin(ogen) consumption during E coli challenge, and thrombin-thrombomodulin, in a baboon model of E coli-induced sepsis, appears to be the predominant activator of TAFI.


Assuntos
Coagulação Sanguínea/fisiologia , Carboxipeptidase B2/metabolismo , Fibrinólise/fisiologia , Trombina/fisiologia , Trombomodulina/fisiologia , Animais , Anticorpos Monoclonais/farmacologia , Carboxipeptidase B2/antagonistas & inibidores , Carboxipeptidase B2/sangue , Carboxipeptidase B2/imunologia , Infecções por Escherichia coli/sangue , Infecções por Escherichia coli/patologia , Produtos de Degradação da Fibrina e do Fibrinogênio/metabolismo , Fibrinogênio/metabolismo , Meia-Vida , Humanos , Testes de Sensibilidade Microbiana , Papio cynocephalus , Sepse/sangue , Sepse/patologia
6.
J Microencapsul ; 24(4): 337-48, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17497387

RESUMO

PRIMARY OBJECTIVE: Antisense oligomers to NF-kappaB (ASO) were incorporated into albumin microspheres to determine if microcapsules containing ASO inhibit pro-inflammatory cytokines to a greater extent than comparable doses of ASO in solution. Phagocytosis of microcapsules and intracellular release of ASO in macrophages was evaluated. RESEARCH DESIGN: Comparable doses of microencapsulated ASO and ASO in solution were evaluated in non-human primates. METHODS: Blood was sampled and stimulated with Escherichia coli endotoxin ex vivo. TNF, IL-1 and IL-6 concentrations were compared for 72 hrs. The intracellular concentration of ASO was measured in macrophages in vitro to evaluate the difference in intracellular penetration of microencapsulated ASO. RESULTS: Microencapsulated ASO produced significantly greater cytokine inhibition at all time points compared to ASO in solution. There were no side effects to ASO in the baboons. Intracellular ASO concentration was 10 fold greater in macrophages using microencapsulation. CONCLUSIONS: Microencapsulated ASO to NF-kappaB is more effective than ASO in solution in pro-inflammatory cytokine inhibition in non-human primates.


Assuntos
Cápsulas , Citocinas/antagonistas & inibidores , Inflamação/fisiopatologia , NF-kappa B/genética , Oligonucleotídeos Antissenso/farmacologia , Análise de Variância , Animais , Emulsões , Macrófagos/efeitos dos fármacos , Macrófagos/fisiologia , Camundongos , Microesferas , Papio , Soroalbumina Bovina
7.
Am J Pathol ; 167(4): 1161-72, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16192650

RESUMO

Endothelium plays a critical role in the pathobiology of sepsis by integrating systemic host responses and local rheological stimuli. We studied the differential expression and activation of tissue factor (TF)-dependent coagulation on linear versus branched arterial segments in a baboon sepsis model. Animals were injected intravenously with lethal doses of Escherichia coli or saline and sacrificed after 2 to 8 hours. Whole-mount arterial segments were stained for TF, TF-pathway inhibitor (TFPI), factor VII (FVII), and markers for endothelial cells (ECs), leukocytes, and platelets, followed by confocal microscopy and image analysis. In septic animals, TF localized preferentially at branches, EC surface, leukocytes, and platelet aggregates and accumulated in large amounts in the subendothelial space. FVII strongly co-localized with TF on ECs and leukocytes but less so with subendothelial TF. TFPI co-localized with TF and FVII on endothelium and leukocytes but not in the subendothelial space. Focal TF increases correlated with fibrin deposition and increased endothelial permeability to plasma proteins. Biochemical analysis confirmed that aortas of septic baboons expressed more TF mRNA and protein than controls. Branched segments contained higher TF protein levels and coagulant activity than equivalent linear areas. These data suggest that site-dependent endothelial heterogeneity and rheological factors contribute to focal procoagulant responses to E. coli.


Assuntos
Artérias/metabolismo , Coagulação Sanguínea , Infecções por Escherichia coli/metabolismo , Choque Séptico/metabolismo , Tromboplastina/metabolismo , Regulação para Cima , Animais , Aorta/metabolismo , Aorta/ultraestrutura , Artérias/anatomia & histologia , Artérias/ultraestrutura , Biomarcadores/sangue , Plaquetas/metabolismo , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Fator VII/metabolismo , Processamento de Imagem Assistida por Computador , Leucócitos/metabolismo , Lipopolissacarídeos/toxicidade , Lipoproteínas/metabolismo , Microscopia Confocal , Modelos Biológicos , Papio , RNA Mensageiro/metabolismo , Choque Séptico/patologia , Choque Séptico/fisiopatologia , Tromboplastina/genética , Tromboplastina/ultraestrutura
8.
Shock ; 22(5): 423-30, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15489634

RESUMO

To monitor and better understand the immunoinflammatory sequelae in sepsis and septic shock, systemic and monocyte-related cytokine responses were evaluated in baboons with experimental peritonitis induced by an E. coli-laden fibrin clot. Despite similar bacterial inocula, considerable interindividual variability in clinical manifestation and outcome of infection was observed. Because monocytes and macrophages are a key component of innate immunity, we hypothesized that early polarization of distinct activation programs in circulating monocytes that culminates in the emergence of either classically (M1) or alternatively (M2) activated monocytes may underlie the observed susceptibility or resistance to infection. To test our hypothesis, we analyzed infection-induced expression of cytokine mRNAs in monocytes isolated from surviving and dead animals. Our data show that resistance to E. coli sepsis may well be associated with a mixed M1/M2 activation state of circulating monocytes, whereas M1 phenotype appeared to be prevailing in monocytes from animals that died. Together with data on systemic cytokine responses, the latter findings indicate that morbidity and mortality of animals with gram-negative sepsis may well result from an overwhelming proinflammatory response. Collectively, our data contribute to a better understanding of cytokine networking in the immunoinflammatory response to microbial infection and suggest M1/M2 immunophenotypic profiling of readily available circulatory monocytes for early prognosis of severe infections.


Assuntos
Monócitos/imunologia , Monócitos/microbiologia , Sepse/sangue , Sepse/diagnóstico , Animais , Linhagem Celular , Células Cultivadas , Citocinas/metabolismo , Primers do DNA/química , Endotoxinas/metabolismo , Escherichia coli/metabolismo , Fibrina/química , Inflamação , Leucócitos Mononucleares/citologia , Camundongos , Monócitos/citologia , Monócitos/metabolismo , Papio , Fenótipo , Reação em Cadeia da Polimerase , Prognóstico , RNA/metabolismo , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sepse/patologia , Fatores de Tempo
9.
Shock ; 20(2): 130-7, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12865656

RESUMO

CD10, also known as neutral endopeptidase or CALLA, is a major metalloproteinase that regulates levels of biologically active peptides that initiate inflammatory, cardiovascular, and neurogenic responses. Relative tissue expression levels of CD10, its peptide substrates, and their receptors constitute the basic regulatory mechanism. Neutrophils contain abundant CD10 and are rapid responders to an inflammatory septic challenge. Expression of neutrophil surface antigens in response to inflammation was studied in the primate model of Escherichia coli-mediated sepsis and in human volunteers injected with lipopolysaccharide (LPS). There was a rapid and profound (up to 95%) reduced baboon neutrophil CD10 expression in response to E. coli injections of 5.71 x 106 CFU/kg to 2.45 x 109 CFU/kg that gradually resolved to preinjection levels. The reduction was both dose and time dependent. Reduced CD10 antigen on mature baboon neutrophils and bands was observed by immunohistochemistry. Human volunteers challenged with 4ng/kg LPS experienced transient chills, nausea, fever, and myalgia. Up to approximately 20% of their neutrophils had reduced CD10 expression, peaking at 2 to 8 h after injection. By 24 h, neutrophil CD10 expression resolved to preinjection levels. In contrast, in both the baboon and human studies, other neutrophil surface antigens were only slightly decreased (CD11a) or increased (CD11b, CD18, CD35, CD66b, and CD63). These data present the novel observation that neutrophil CD10 expression decreases significantly in response to in vivo inflammatory challenge. This decrease appears to be unique to CD10 and may contribute to a reduced regulation of bioactive peptides released in response to inflammatory challenge.


Assuntos
Escherichia coli/metabolismo , Lipopolissacarídeos/metabolismo , Neprilisina/biossíntese , Neutrófilos/metabolismo , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais/metabolismo , Relação Dose-Resposta a Droga , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Inflamação , Neprilisina/metabolismo , Papio , Sepse/imunologia , Células-Tronco , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA