Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Vasc Surg ; 68(6S): 209S-221S.e2, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-29395422

RESUMO

OBJECTIVE: Ischemia-reperfusion (I/R) injury is a major clinical problem linked to vascular surgery. Currently, no drugs to prevent or to treat I/R injury are approved for clinical use. C1 inhibitor (C1 INH) is known to reduce activation of the plasma cascade systems that are involved in the pathophysiologic process of I/R injury. The aim of this study was therefore to investigate the effect of C1 INH on complement deposition and endothelial cell activation in a rat model of hind limb I/R injury. METHODS: Male Wistar rats (wild type, bred at the central animal facility, University of Bern), weighing 250 to 320 g, were used. The rats underwent 2-hour ischemia and 24-hour reperfusion by unilateral clamping of the femoral artery and additional use of a tourniquet. Five groups were divided according to intravenous treatment 5 minutes before ischemia: 50 IU/kg C1 INH (n = 5); 100 IU/kg C1 INH (n = 7); vehicle control (n = 5); nontreated control (n = 7); and normal, healthy control without intervention (n = 4). At the end, muscle edema, tissue viability, and histologic features were assessed. Deposition of immunoglobulin M, C1r, C4d, and fibrin and expression of plasminogen activator inhibitor 1, heparan sulfate (HS), E-selectin, and vascular cell adhesion molecule 1 were evaluated by fluorescence staining. In addition, high-mobility group box 1 protein was measured in plasma. RESULTS: Edema formation was reduced by C1 INH at two dosages, mirrored by improved histologic injury scores and preserved muscle viability. Deposition of immunoglobulin M, C4d, and fibrin was significantly decreased by 100 IU/kg C1 INH compared with nontreated controls. Pretreatment with 100 IU/kg C1 INH also significantly reduced HS shedding and expression of plasminogen activator inhibitor 1 as well as plasma levels of high-mobility group box 1 protein. CONCLUSIONS: Pretreatment with both 50 and 100 IU/kg C1 INH attenuated reperfusion injury of rat hind limbs. Pretreatment with 100 IU/kg also preserved the endothelial HS layer as well as the natural, profibrinolytic phenotype of the endothelium. Prevention of endothelial cell activation by C1 INH may therefore be a promising strategy to prevent I/R injury in the clinical setting of peripheral vascular diseases and elective surgery on extremities.


Assuntos
Ativação do Complemento/efeitos dos fármacos , Proteína Inibidora do Complemento C1/farmacologia , Inativadores do Complemento/farmacologia , Células Endoteliais/efeitos dos fármacos , Músculo Esquelético/irrigação sanguínea , Músculo Esquelético/efeitos dos fármacos , Traumatismo por Reperfusão/prevenção & controle , Animais , Complemento C1r/metabolismo , Complemento C4b/metabolismo , Modelos Animais de Doenças , Selectina E/metabolismo , Edema/imunologia , Edema/metabolismo , Edema/patologia , Edema/prevenção & controle , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Fibrina/metabolismo , Proteína HMGB1/metabolismo , Heparitina Sulfato/metabolismo , Membro Posterior , Imunoglobulina M/metabolismo , Masculino , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Fragmentos de Peptídeos/metabolismo , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Ratos Wistar , Traumatismo por Reperfusão/imunologia , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Sobrevivência de Tecidos/efeitos dos fármacos , Molécula 1 de Adesão de Célula Vascular/metabolismo
2.
Am J Respir Crit Care Med ; 196(2): 186-199, 2017 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-28005404

RESUMO

RATIONALE: Acute respiratory distress syndrome is characterized by alveolar epithelial cell injury, edema formation, and intraalveolar contact phase activation. OBJECTIVES: To explore whether C1 esterase inhibitor (C1INH), an endogenous inhibitor of the contact phase, may protect from lung injury in vivo and to decipher the possible underlying mechanisms mediating protection. METHODS: The ability of C1INH to control the inflammatory processes was studied in vitro and in vivo. MEASUREMENTS AND MAIN RESULTS: Here, we demonstrate that application of C1INH alleviates bleomycin-induced lung injury via direct interaction with extracellular histones. In vitro, C1INH was found to bind all histone types. Interaction with histones was independent of its protease inhibitory activity, as demonstrated by the use of reactive-center-cleaved C1INH, but dependent on its glycosylation status. C1INH sialylated-N- and -O-glycans were not only essential for its interaction with histones but also to protect against histone-induced cell death. In vivo, histone-C1INH complexes were detected in bronchoalveolar lavage fluid from patients with acute respiratory distress syndrome and multiple models of lung injury. Furthermore, reactive-center-cleaved C1INH attenuated pulmonary damage evoked by intravenous histone instillation. CONCLUSIONS: Collectively, C1INH administration provides a new therapeutic option for disorders associated with histone release.


Assuntos
Proteína Inibidora do Complemento C1/farmacologia , Histonas/metabolismo , Lesão Pulmonar/prevenção & controle , Síndrome do Desconforto Respiratório/fisiopatologia , Animais , Líquido da Lavagem Broncoalveolar , Proteína Inibidora do Complemento C1/metabolismo , Modelos Animais de Doenças , Humanos , Pulmão/metabolismo , Pulmão/fisiopatologia , Lesão Pulmonar/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL
3.
Thromb Haemost ; 117(1): 176-187, 2017 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-27786338

RESUMO

Atherosclerosis is considered a chronic inflammatory disease of the vessel wall. Coagulation pathways and immune responses contribute to disease development. The role of coagulation factor XII (FXII) in vascular inflammation, however, remains controversial. We here investigated the function of FXII in atherosclerosis using apolipoprotein E and FXII-deficient (F12-/-Apoe-/-) mice. Compared to F12+/+Apoe-/- controls, atherosclerotic lesion formation was reduced in F12-/-Apoe-/- mice. This was associated with a decrease in serum interleukin (IL)-1ß and IL-12 levels and reduced expression of pro-inflammatory cytokines in the aorta in atherosclerotic F12-/-Apoe-/- mice, as well as diminished Th1-cell differentiation in the aorta, blood, and lymphoid organs. No changes in circulating bradykinin, thrombin-antithrombin-complexes or plasminogen were observed. Mechanistically, activated FXII (FXIIa) was revealed to directly induce bone marrow-derived macrophages to secrete pro-inflammatory cytokines, including tumour necrosis factor-α, IL-1ß, IL-12, and IL-6. Exposure of bone marrow-derived antigen presenting cells to FXIIa similarly induced pro-inflammatory cytokines, and an enhanced capacity to trigger antigen-specific interferon γ-production in CD4+ T cells. Notably, bone-marrow derived macrophages were capable of directly activating FXII. Moreover, the induction of cytokine expression by FXIIa in macrophages occurred independently of FXII protease enzymatic activity and was decreased upon phospholipase C treatment, suggesting urokinase-type plasminogen activator receptor (uPAR) to confer FXIIa-induced cell signalling. These data reveal FXII to play an important role in atherosclerotic lesion formation by functioning as a strong inducer of pro-inflammatory cytokines in antigen-presenting cells. Targeting of FXII may thus be a promising approach for treating cardiovascular disease.


Assuntos
Células Apresentadoras de Antígenos/metabolismo , Doenças da Aorta/metabolismo , Aterosclerose/metabolismo , Citocinas/metabolismo , Deficiência do Fator XII/metabolismo , Fator XII/metabolismo , Mediadores da Inflamação/metabolismo , Macrófagos/metabolismo , Animais , Células Apresentadoras de Antígenos/imunologia , Doenças da Aorta/sangue , Doenças da Aorta/genética , Doenças da Aorta/imunologia , Aterosclerose/sangue , Aterosclerose/genética , Aterosclerose/imunologia , Proliferação de Células , Citocinas/imunologia , Modelos Animais de Doenças , Fator XII/genética , Deficiência do Fator XII/sangue , Deficiência do Fator XII/genética , Deficiência do Fator XII/imunologia , Fator XIIa/genética , Fator XIIa/metabolismo , Predisposição Genética para Doença , Mediadores da Inflamação/imunologia , Ativação Linfocitária , Macrófagos/imunologia , Camundongos Endogâmicos C57BL , Camundongos Knockout para ApoE , Fenótipo , Placa Aterosclerótica , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Células Th1/imunologia , Células Th1/metabolismo , Fatores de Tempo
4.
PLoS One ; 8(5): e63162, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23696796

RESUMO

Intravenous Immunoglobulin (IVIG) has been proposed as a potential therapeutic for Alzheimer's disease (AD) and its efficacy is currently being tested in mild-to-moderate AD. Earlier studies reported the presence of anti-amyloid beta (Aß) antibodies in IVIG. These observations led to clinical studies investigating the potential role of IVIG as a therapeutic agent in AD. Also, IVIG is known to mediate beneficial effects in chronic inflammatory and autoimmune conditions by interfering with various pathological processes. Therefore, we investigated the effects of IVIG and purified polyclonal Aß-specific antibodies (pAbs-Aß) on aggregation, toxicity and phagocytosis of Aß in vitro, thus elucidating some of the potential mechanisms of action of IVIG in AD patients. We report that both IVIG and pAbs-Aß specifically bound to Aß and inhibited its aggregation in a dose-dependent manner as measured by Thioflavin T assay. Additionally, IVIG and the purified pAbs-Aß inhibited Aß-induced neurotoxicity in the SH-SY5Y human neuroblastoma cell line and prevented Aß binding to rat primary cortical neurons. Interestingly, IVIG and pAbs-Aß also increased the number of phagocytosing cells as well as the amount of phagocytosed fibrillar Aß by BV-2 microglia. Phagocytosis of Aß depended on receptor-mediated endocytosis and was accompanied by upregulation of CD11b expression. Importantly, we could also show that Privigen dose-dependently reversed Aß-mediated LTP inhibition in mouse hippocampal slices. Therefore, our in vitro results suggest that IVIG may have an impact on different processes involved in AD pathogenesis, thereby promoting further understanding of the effects of IVIG observed in clinical studies.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Imunoglobulinas/metabolismo , Microglia/citologia , Microglia/metabolismo , Fagocitose/fisiologia , Peptídeos beta-Amiloides/genética , Animais , Antígeno CD11b/metabolismo , Linhagem Celular Tumoral , Cromatografia de Afinidade , Ensaio de Imunoadsorção Enzimática , Humanos , Imunoglobulinas/genética , Imuno-Histoquímica , Camundongos , Microscopia de Força Atômica
5.
Neurobiol Dis ; 31(3): 342-54, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18632280

RESUMO

Neural transplantation has been investigated experimentally and clinically for the purpose of developing new treatment options for intractable epilepsy. In the present study we assessed the anticonvulsant efficacy and safety of bilateral allotransplantation of genetically engineered striatal GABAergic rat cell lines into the substantia nigra pars reticulata (SNr). Rats with previously-established seizures, induced by amygdala kindling, were used as a model of temporal lobe epilepsy. Three cell lines were transplanted: (1) immortalized GABAergic cells (M213-2O) derived from embryonic rat striatum; (2) M213-2O cells (CL4) transfected with human GAD67 cDNA to obtain higher GABA synthesis than the parent cell line; and (3) control cells (121-1I), also derived from embryonic rat striatum, but which did not show GAD expression. A second control group received injections of medium alone. Transplantation of M213-2O cells into the SNr of kindled rats resulted in significant but transient anticonvulsant effects. Neither control cells nor medium induced anticonvulsant effects. Strong tissue reactions were, however, induced in the host brain of kindled but not of non-kindled rats, and only in animals that received grafts of genetically modified CL4 cells. These tissue reactions included graft rejection, massive infiltration of inflammatory immune cells, and gliosis. The anticonvulsant effect of M213-2O cells emphasizes the feasibility of local manipulations of seizures by intranigral transplantation of GABA-producing cells. On the other hand, the present data suggest that kindling-induced activation of microglia in the SNr can enhance immune reactions to transplanted cells. In this case, under conditions of further immunological stimulation by CL4 cells, transfected with a human cDNA, substantial immune reactions occurred. Thus, it appears that the condition of the host brain and the production of foreign proteins by transplanted cells have to be considered in estimating the risks of rejection of transplants into the brain.


Assuntos
Transplante de Tecido Encefálico/métodos , Epilepsia/metabolismo , Epilepsia/cirurgia , Substância Negra/metabolismo , Substância Negra/cirurgia , Ácido gama-Aminobutírico/biossíntese , Animais , Transplante de Tecido Encefálico/efeitos adversos , Linhagem Celular Transformada , Modelos Animais de Doenças , Epilepsia/fisiopatologia , Feminino , Terapia Genética/métodos , Glutamato Descarboxilase/genética , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/metabolismo , Rejeição de Enxerto/prevenção & controle , Humanos , Excitação Neurológica/metabolismo , Microglia/imunologia , Inibição Neural/fisiologia , Neurônios/citologia , Neurônios/metabolismo , Neurônios/transplante , Ratos , Ratos Wistar , Medição de Risco , Transplante de Células-Tronco/efeitos adversos , Transplante de Células-Tronco/métodos , Substância Negra/fisiopatologia , Transfecção/métodos , Resultado do Tratamento , Regulação para Cima/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA