Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Clin Immunol ; 263: 110207, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38608995

RESUMEN

Behçet's disease (BD) is an autoinflammatory disease with multifactorial and polygenic etiology, potentially involving arteries and veins of any size resulting in variable vessel vasculitis. We report a case of an Iranian male who presented with porto-sinusoidal vascular disorder due to venous vasculitis as initial manifestation of BD. Despite immunosuppression, anticoagulation and venous recanalization, he subsequently developed severe nephrotic-range proteinuria mimicking a primary renal disease which was completely and immediately ameliorated by stenting of the vena cava. This demonstrates that the proteinuria was caused by increased intraglomerular pressure due to venous outflow obstruction as a consequence of venous vasculitis. To our knowledge, this is the first report of massive proteinuria caused by venous obstruction of the caval vein in the context of BD. Altogether, this case demonstrates the extensive spectrum of vascular disease in BD.


Asunto(s)
Síndrome de Behçet , Proteinuria , Humanos , Masculino , Síndrome de Behçet/complicaciones , Proteinuria/etiología , Vasculitis/etiología
2.
Virchows Arch ; 481(2): 245-252, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35513609

RESUMEN

Rectal suction biopsy (RSB) is a gold standard for diagnosing Hirschsprung disease (HD). Calretinin staining of RSB is increasingly used by experienced pathologists due to non-complex examination and comparable diagnostic accuracy with acetylcholinesterase (AChE). However, the diagnostic accuracy of calretinin examined by unexperienced pathologists remains to be elucidated. Therefore, we aim to compare diagnostic accuracy of calretinin with AChE on RSB for diagnosing HD when examined by unexperienced pathologists. We prospectively analyzed sections from RSB stained with AChE + HE and calretinin. Blinded examination was done by five unexperienced pathologists (pathology residents) and three experienced pathologists (senior pediatric gastro-enterology pathologists) assessing for the presence of HD. Cases for the study included ones proven to be HD on resection specimens and cases without HD. Diagnostic accuracy was determined calculating area under the curve (AUC), sensitivity, specificity, likelihood ratio, and posttest probability. Fleiss' kappa analysis was performed to assess interobserver agreement between reviewers. Eleven of 18 included patients (61%) were diagnosed with HD. Comparing the diagnostic accuracy of unexperienced pathologists, calretinin versus AChE + HE showed sensitivity of 80.0% versus 74.5% and specificity of 100% versus 65.4%, AUC of 0.87 (0.78-0.96) versus 0.59 (0.45-0.72). Unexperienced pathologists showed substantial agreement with calretinin (kappa 0.72 [0.61-0.84]) and fair agreement with AChE + HE (kappa 0.34 [0.23-0.44]). We found calretinin having higher diagnostic accuracy in diagnosing HD compared to AChE + HE when examined by unexperienced pathologists. Therefore, we recommend to use calretinin as the standard technique for staining RSB in diagnosing HD.


Asunto(s)
Enfermedad de Hirschsprung , Acetilcolinesterasa/análisis , Acetilcolinesterasa/metabolismo , Biopsia/métodos , Calbindina 2/análisis , Niño , Enfermedad de Hirschsprung/diagnóstico , Humanos , Lactante , Patólogos , Recto/patología , Coloración y Etiquetado , Succión
3.
J Thromb Haemost ; 15(4): 744-757, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28092405

RESUMEN

Essentials Immunity and coagulation are linked during sepsis but the role of thrombin is not fully elucidated. We investigated the effect of thrombin inhibition on murine Klebsiella pneumosepsis outcome. Thrombin is crucial for survival and limiting bacterial growth in pneumonia derived sepsis. Thrombin improves host defense via fibrin and enhancement of platelet-neutrophil interactions. SUMMARY: Background Innate immunity and coagulation are closely linked during sepsis. Their interaction can be detrimental to the outcome because of microvascular failure but can also enhance host defense. The role of thrombin therein has not been fully elucidated. Objective We aimed to investigate the contribution of thrombin to the host response during pneumonia-derived sepsis. Methods Mice treated with the specific thrombin inhibitor dabigatran or control chow were infected with the common human sepsis pathogen Klebsiella (K.) pneumoniae via the airways. In subsequent infection experiments, mice were additionally treated with ancrod to deplete fibrinogen. Ex vivo Klebsiella growth was assessed by incubating human whole blood or specific blood components in various conditions with Klebsiella. Results Thrombin inhibition by dabigatran enhanced bacterial outgrowth and spreading, and accelerated mortality. Thrombin inhibition did not influence neutrophil recruitment to the lung or activation or neutrophil extracellular trap formation. Dabigatran reduced D-dimer formation and fibrin deposition in the lung. Fibrin depletion also enhanced bacterial outgrowth and spreading, and thrombin inhibition had no additional effect. Both thrombin and fibrin polymerization inhibited ex vivo Klebsiella outgrowth in human whole blood, which was neutrophil dependent, and the effect of thrombin required the presence of platelets and platelet protease activated receptor-1. In vivo thrombin inhibition reduced platelet-neutrophil complex formation and endothelial cell activation, but did not prevent sepsis-induced thrombocytopenia or organ damage. Conclusions These results suggest that thrombin plays an important role in protective immunity during pneumonia-derived sepsis by fibrin polymerization and enhancement of platelet-neutrophil interactions.


Asunto(s)
Plaquetas/citología , Fibrina/química , Neutrófilos/citología , Neumonía Bacteriana/inmunología , Sepsis/inmunología , Trombina/inmunología , Animales , Coagulación Sanguínea , Comunicación Celular , Dabigatrán/administración & dosificación , Trampas Extracelulares , Femenino , Fibrinógeno/química , Citometría de Flujo , Humanos , Sistema Inmunológico , Inmunidad Innata , Infecciones por Klebsiella/inmunología , Klebsiella pneumoniae , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Microcirculación , Sepsis/microbiología
4.
Clin Exp Immunol ; 187(2): 204-212, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27625307

RESUMEN

Asthma is a highly prevalent chronic allergic inflammatory disease of the airways affecting people worldwide. House dust mite (HDM) is the most common allergen implicated in human allergic asthma. HDM-induced allergic responses are thought to depend upon activation of pathways involving Toll-like receptors and their adaptor protein myeloid differentiation factor 88 (MyD88). We sought here to determine the role of MyD88 in myeloid and type II lung epithelial cells in the development of asthma-like allergic disease using a mouse model. Repeated exposure to HDM caused allergic responses in control mice characterized by influx of eosinophils into the bronchoalveolar space and lung tissue, lung pathology and mucus production and protein leak into bronchoalveolar lavage fluid. All these responses were abrogated in mice with a general deficiency of MyD88 but unaltered in mice with MyD88 deficiency, specifically in myeloid or type II lung epithelial cells. We conclude that cells other than myeloid or type II lung epithelial cells are responsible for MyD88-dependent HDM-induced allergic airway inflammation.


Asunto(s)
Asma/inmunología , Células Epiteliales/inmunología , Hipersensibilidad/inmunología , Células Mieloides/fisiología , Factor 88 de Diferenciación Mieloide/metabolismo , Neumonía/inmunología , Pyroglyphidae/inmunología , Animales , Antígenos Dermatofagoides/inmunología , Movimiento Celular , Células Epiteliales/patología , Humanos , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética
5.
J Thromb Haemost ; 14(6): 1171-82, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26990852

RESUMEN

UNLABELLED: Essentials Endothelial protein C receptor (EPCR) promotes diabetic nephropathy (DN) outcome improvement. Renal expression and shedding of EPCR were measured in diabetic patients with or without DN. Inhibition of metalloproteinase-driven EPCR shedding restored glomerular endothelium phenotype. EPCR shedding through metalloproteinase ADAM17 contributes to the worsening of DN. SUMMARY: Background Diabetic nephropathy (DN) represents the leading cause of end-stage renal disease. The endothelial protein C receptor (EPCR) and its ligand (activated protein C) have been shown to ameliorate the phenotype of DN in mice. EPCR activity can be regulated by proteolytic cleavage involving ADAMs, yielding a soluble form of EPCR (sEPCR). Objective To characterize the renal expression and shedding of EPCR during DN. Methods EPCR levels were measured in plasma, urine and biopsy samples of diabetic patients with (n = 73) or without (n = 63) DN. ADAM-induced cleavage of EPCR was investigated in vitro with a human glomerular endothelium cell line. Results DN patients showed higher plasma and urinary levels of sEPCR than diabetic controls (112.2 versus 135.2 ng mL(-1) and 94.35 versus 140.6 ng mL(-1) , respectively). Accordingly, glomerular endothelial EPCR expression was markedly reduced in patients with DN, and this was associated with increased glomerular expression of ADAM-17 and ADAM-10. In vitro, EPCR shedding was induced by incubation of glomerular endothelium in high-glucose medium, and this shedding was suppressed by ADAM-17 inhibition or silencing, which led to improved vascular endothelial cadherin (VE-cadherin) expression and reduced mRNA expression of transforming growth factor (TGF)-ß. In addition, EPCR silencing led to minor effects on VE-cadherin but to a significant increase in TGF-ß mRNA expression. Conclusion Inhibition of ADAM-driven glomerular EPCR shedding restored the endothelial phenotype of glomerular endothelium, whereas EPCR silencing led to enhanced expression of TGF-ß, a marker of endothelial-mesenchymal transition. These findings demonstrate that EPCR shedding driven by ADAMs contributes to the worsening of DN.


Asunto(s)
Nefropatías Diabéticas/metabolismo , Receptor de Proteína C Endotelial/metabolismo , Riñón/metabolismo , Proteína ADAM10/metabolismo , Proteína ADAM17/metabolismo , Anciano , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Biopsia , Línea Celular , Diabetes Mellitus/sangre , Diabetes Mellitus/patología , Diabetes Mellitus/orina , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Nefropatías Diabéticas/patología , Endotelio/patología , Femenino , Silenciador del Gen , Humanos , Glomérulos Renales/metabolismo , Ligandos , Masculino , Proteínas de la Membrana/metabolismo , Metaloproteasas/metabolismo , Persona de Mediana Edad , Fenotipo , ARN Interferente Pequeño/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
6.
Mucosal Immunol ; 9(4): 960-73, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26530135

RESUMEN

MiR-511-3p is embedded in intron 5 of the CD206/MRC1 gene Mrc1, expressed by macrophage and dendritic cell populations. CD206 and miR-511-3p expression are co-regulated, and their contribution to intestinal inflammation is unclear. We investigated their roles in intestinal inflammation in both mouse and human systems. Colons of CD206-deficient mice displayed normal numbers of monocytes, macrophage, and dendritic cells. In experimental colitis, CD206-deficient mice had attenuated inflammation compared with wild-type (WT) mice. However, neither a CD206 antagonist nor a blocking antibody reproduced this phenotype, suggesting that CD206 was not involved in this response. Macrophages isolated from CD206-deficient mice had reduced levels of miR-511-3p and Tlr4 compared with WT, which was associated with reduced pro-inflammatory cytokine production upon lipopolysaccharides (LPS) and fecal supernatant stimulation. Macrophages overexpressing miR-511-3p showed 50% increase of Tlr4 mRNA, whereas knockdown of miR-511-3p reduced Tlr4 mRNA levels by 60%, compared with scrambled microRNA (miRNA)-transduced cells. Response to anti-tumor necrosis factor (TNF) treatment has been associated with elevated macrophage CD206 expression in the mucosa. However, in colon biopsies no statistically significant change in miR-511-3p was detected. Taken together, our data show that miR-511-3p controls macrophage-mediated microbial responses and is involved in the regulation of intestinal inflammation.


Asunto(s)
Colitis/inmunología , Colon/inmunología , Macrófagos/inmunología , Glicoproteínas de Membrana/genética , MicroARNs/genética , Receptores de Superficie Celular/genética , Animales , Células Cultivadas , Colitis/inducido químicamente , Sulfato de Dextran , Femenino , Regulación de la Expresión Génica , Humanos , Lipopolisacáridos/inmunología , Masculino , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Superficie Celular/metabolismo , Receptores Inmunológicos , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
7.
Allergy ; 70(12): 1531-44, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26222912

RESUMEN

BACKGROUND: Asthma is an inflammatory disease that involves airway hyper-responsiveness and mucus hypersecretion. The LIM-only protein FHL2 is a crucial modulator of multiple signal transduction pathways and functions as a scaffold in specific protein-protein interactions. OBJECTIVE: We sought to investigate the role of FHL2 in airway inflammation. METHODS: Allergic airway inflammation was induced in WT and FHL2-knock out (FHL2-KO) mice with ovalbumin (OVA). Lung tissue, bronchoalveolar lavage fluid (BALF) and draining lymph node cells were analysed for inflammation. FHL2 loss and gain of function studies were performed in lung epithelial cells. RESULTS: FHL2-deficient mice challenged with OVA show significantly reduced airway inflammation as evidenced by reduced infiltration of inflammatory cells including eosinophils, dendritic cells, B cells and T cells. Furthermore, mucus production was decreased in FHL2-KO mice. In BALF, the levels of IL-5, IL-13, eotaxin-1 and eotaxin-2 were significantly lower in FHL2-KO mice. In addition, draining lymph node cells from FHL2-KO mice show reduced levels of IL-5 and IL-13. Consistent with this, OVA-specific serum IgG and IgE levels were reduced in FHL2-KO mice. We also found that phosphorylation of ERK1/2 is markedly attenuated in FHL2-KO lung. Knock-down of FHL2 in human lung epithelial cells resulted in a striking decrease in ERK1/2 phosphorylation and mRNA levels of inflammatory cytokines and MUC5AC, whereas FHL2 overexpression exhibited opposite effects. Finally, the SNP rs4851765 shows an association with the severity of bronchial hyper-responsiveness. CONCLUSION: These results highlight functional involvement of FHL2 in airway inflammation and identify FHL2 as a novel gene associated with asthma severity in human.


Asunto(s)
Asma/genética , Proteínas con Homeodominio LIM/metabolismo , Proteínas Musculares/metabolismo , Neumonía/genética , Hipersensibilidad Respiratoria/genética , Factores de Transcripción/metabolismo , Animales , Asma/metabolismo , Western Blotting , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad/genética , Genotipo , Humanos , Proteínas con Homeodominio LIM/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Musculares/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Neumonía/metabolismo , Reacción en Cadena de la Polimerasa , Polimorfismo de Nucleótido Simple , Hipersensibilidad Respiratoria/metabolismo , Factores de Transcripción/genética
8.
J Thromb Haemost ; 13(9): 1709-20, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26178922

RESUMEN

BACKGROUND: Beside their role in hemostasis, platelets serve as sentinel cells in host defense during infection. In sepsis, platelets have been implicated in both beneficial (antibacterial) and detrimental responses (thrombosis and organ damage). Toll-like receptors and their common adaptor, myeloid differentiation factor 88 (MyD88), are essential for pathogen recognition and protective immunity. Platelets express functional Toll-like receptors and MyD88, which participate in platelet responsiveness to bacterial agonists. OBJECTIVE: Considering the pivotal involvement of platelets and MyD88 in the host response to bacteria, we studied the role of platelet MyD88 in gram-negative sepsis using intravenous and airway infections with the common human sepsis pathogen Klebsiella pneumoniae. METHODS: Platelet-specific Myd88(-/-) mice were generated by crossing mice with a conditional Myd88 flox allele with mice expressing Cre recombinase controlled by the platelet factor 4 promoter. In a reverse approach, full Myd88(-/-) mice were transfused with wild-type platelets. RESULTS: In both settings, platelet MyD88 did not impact on bacterial growth or dissemination. In addition, platelet MyD88 did not influence hallmark sepsis responses such as thrombocytopenia, coagulation or endothelial activation, or distant organ injury. Platelet MyD88 played no role in lung pathology during pneumonia-derived sepsis. CONCLUSION: Despite known literature, platelet MyD88-dependent TLR signaling does not contribute to the host response during gram-negative sepsis.


Asunto(s)
Plaquetas/inmunología , Infecciones por Klebsiella/inmunología , Klebsiella pneumoniae/inmunología , Factor 88 de Diferenciación Mieloide/fisiología , Sepsis/inmunología , Receptores Toll-Like/sangre , Animales , Bacteriemia/complicaciones , Bacteriemia/inmunología , Bacteriemia/microbiología , Carga Bacteriana , Coagulación Sanguínea , Quimiocina CCL2/sangre , Endotelio Vascular/fisiopatología , Trampas Extracelulares , Femenino , Infecciones por Klebsiella/sangre , Infecciones por Klebsiella/terapia , Hígado/patología , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/deficiencia , Transfusión de Plaquetas , Neumonía Bacteriana/sangre , Neumonía Bacteriana/complicaciones , Neumonía Bacteriana/inmunología , Neumonía Bacteriana/microbiología , Neumonía Bacteriana/patología , Sepsis/sangre , Sepsis/etiología , Sepsis/terapia , Método Simple Ciego , Bazo/patología , Factor de Necrosis Tumoral alfa/análisis
9.
J Thromb Haemost ; 13(6): 1128-38, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25773400

RESUMEN

BACKGROUND: Sepsis is associated with activation of platelets and endothelial cells accompanied by enhanced P-selectin surface expression. Both platelet- and endothelial P-selectin have been associated with leukocyte recruitment and induction of inflammatory alterations. Klebsiella (K.) pneumoniae is a common human sepsis pathogen, particularly in the context of pneumonia. METHODS: Wild-type (WT) and P-selectin-deficient (Selp(-/-) ) mice or bone marrow chimeric mice were infected with K. pneumoniae via the airways to induce pneumosepsis. Mice were sacrificed during early (12 h after infection) or late-stage (44 h) sepsis for analyses, or followed in a survival study. RESULTS: Selp(-/-) mice displayed 10-1000-fold higher bacterial burdens in the lungs, blood and distant organs during late-stage sepsis. P-selectin deficiency did not influence leukocyte recruitment to the lungs, but was associated with decreased platelet-monocyte complexes and increased cytokine release. Bone marrow transfer studies revealed a role for both platelet and endothelial cell P-selectin as mice deficient in platelet or endothelial cell P-selectin displayed an intermediate phenotype in bacterial loads and survival compared with full wild-type or full knockout control mice. CONCLUSION: Both platelet and endothelial cell P-selectin contribute to host defense during Klebsiella pneumosepsis.


Asunto(s)
Plaquetas/metabolismo , Células Endoteliales/metabolismo , Infecciones por Klebsiella/metabolismo , Klebsiella pneumoniae/patogenicidad , Selectina-P/metabolismo , Neumonía Bacteriana/metabolismo , Sepsis/metabolismo , Animales , Carga Bacteriana , Coagulación Sanguínea , Plaquetas/inmunología , Plaquetas/microbiología , Trasplante de Médula Ósea , Quimiotaxis de Leucocito , Citocinas/sangre , Modelos Animales de Enfermedad , Células Endoteliales/inmunología , Células Endoteliales/microbiología , Interacciones Huésped-Patógeno , Inmunidad Innata , Mediadores de Inflamación/sangre , Infecciones por Klebsiella/genética , Infecciones por Klebsiella/inmunología , Infecciones por Klebsiella/microbiología , Klebsiella pneumoniae/crecimiento & desarrollo , Klebsiella pneumoniae/inmunología , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/microbiología , Ratones Endogámicos C57BL , Ratones Noqueados , Selectina-P/genética , Activación Plaquetaria , Neumonía Bacteriana/genética , Neumonía Bacteriana/inmunología , Neumonía Bacteriana/microbiología , Factores Protectores , Sepsis/genética , Sepsis/inmunología , Sepsis/microbiología , Transducción de Señal , Factores de Tiempo
10.
J Thromb Haemost ; 12(6): 943-7, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24679129

RESUMEN

BACKGROUND: A common complication after aneurysmal subarachnoid hemorrhage (SAH) is delayed cerebral ischemia (DCI), which is associated with vasospasm and other mechanisms such as microthrombosis. ADAMTS-13 activity plays a role in the prevention of thrombus formation in the cerebral microvasculature. Previously, we observed that patients with DCI have lower levels of ADAMTS-13. OBJECTIVES: To examine whether recombinant human ADAMTS-13 (rADAMTS-13) reduces cerebral microthrombus formation and brain injury in an experimental mouse model of SAH including wild-type and ADAMTS-13(-/-) mice. METHODS: Experimental SAH was induced with the prechiasmatic blood injection model. The following experimental groups were investigated: (i) C57BL/6J mice (n = 10); (ii) C57BL/6J mice (n = 10) treated with rADAMTS-13 20 min after SAH; (iii) ADAMTS-13(-/-) mice (n = 10); and (iv) ADAMTS-13(-/-) mice (n = 10) treated with rADAMTS-13 20 min after SAH. Mice were killed at 48 h. Results are presented as means with standard errors of the mean. RESULTS: Infusion with rADAMTS-13 reduced the extent of microthrombosis by ~ 50% in both wild-type mice (mean fibrinogen area: 0.28% ± 0.09% vs. 0.15% ± 0.04%; P = 0.20) and ADAMTS-13(-/-) mice (mean fibrinogen area: 0.32% ± 0.05% vs. 0.16% ± 0.03%; P = 0.016). In addition, rADAMTS-13 reduced brain injury by > 60% in both wild-type mice (mean microglia area: 0.65% ± 0.18% vs. 0.18% ± 0.04%; P = 0.013) and ADAMTS-13(-/-) mice (mean microglia area: 1.24% ± 0.36% vs. 0.42% ± 0.13%; P = 0.077). CONCLUSIONS: Our results support the further study of rADAMTS-13 as a treatment option for the prevention of microthrombosis and brain injury after SAH.


Asunto(s)
Proteínas ADAM/farmacología , Lesiones Encefálicas/prevención & control , Isquemia Encefálica/prevención & control , Encéfalo/efectos de los fármacos , Trombosis Intracraneal/prevención & control , Fármacos Neuroprotectores/farmacología , Hemorragia Subaracnoidea/complicaciones , Proteína ADAMTS13 , Animales , Encéfalo/patología , Lesiones Encefálicas/diagnóstico , Lesiones Encefálicas/etiología , Isquemia Encefálica/diagnóstico , Isquemia Encefálica/etiología , Citoprotección , Modelos Animales de Enfermedad , Humanos , Trombosis Intracraneal/diagnóstico , Trombosis Intracraneal/etiología , Metaloendopeptidasas/deficiencia , Metaloendopeptidasas/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Recombinantes/farmacología , Factores de Tiempo
11.
Cell Biochem Biophys ; 70(2): 795-803, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24760631

RESUMEN

Mechanical ventilation has the potential to cause lung injury, and the role of complement activation herein is uncertain. We hypothesized that inhibition of the complement cascade by administration of plasma-derived human C1-esterase inhibitor (C1-INH) prevents ventilation-induced pulmonary complement activation, and as such attenuates lung inflammation and lung injury in a rat model of Streptococcus pneumoniae pneumonia. Forty hours after intratracheal challenge with S. pneumoniae causing pneumonia rats were subjected to ventilation with lower tidal volumes and positive end-expiratory pressure (PEEP) or high tidal volumes without PEEP, after an intravenous bolus of C1-INH (200 U/kg) or placebo (saline). After 4 h of ventilation blood, broncho-alveolar lavage fluid and lung tissue were collected. Non-ventilated rats with S. pneumoniae pneumonia served as controls. While ventilation with lower tidal volumes and PEEP slightly amplified pneumonia-induced complement activation in the lungs, ventilation with higher tidal volumes without PEEP augmented local complement activation more strongly. Systemic pre-treatment with C1-INH, however, failed to alter ventilation-induced complement activation with both ventilation strategies. In accordance, lung inflammation and lung injury were not affected by pre-treatment with C1-INH, neither in rats ventilated with lower tidal volumes and PEEP, nor rats ventilated with high tidal volumes without PEEP. Ventilation augments pulmonary complement activation in a rat model of S. pneumoniae pneumonia. Systemic administration of C1-INH, however, does not attenuate ventilation-induced complement activation, lung inflammation, and lung injury.


Asunto(s)
Activación de Complemento/efectos de los fármacos , Proteína Inhibidora del Complemento C1/farmacología , Pulmón/efectos de los fármacos , Pulmón/inmunología , Neumonía/terapia , Respiración Artificial/efectos adversos , Streptococcus pneumoniae/fisiología , Animales , Modelos Animales de Enfermedad , Humanos , Pulmón/microbiología , Masculino , Ratas , Ratas Wistar , Factores de Tiempo , Lesión Pulmonar Inducida por Ventilación Mecánica/etiología , Lesión Pulmonar Inducida por Ventilación Mecánica/inmunología , Lesión Pulmonar Inducida por Ventilación Mecánica/prevención & control
12.
Vox Sang ; 107(1): 71-5, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24372323

RESUMEN

BACKGROUND AND OBJECTIVE: Transfusion-related acute lung injury (TRALI) is the leading cause of transfusion-related morbidity and mortality. Specific therapy is lacking. We assessed whether C1-inhibitor attenuates lung injury in a 'two-hit' TRALI model. METHODS: Mice were primed with lipopolysaccharide, subsequently TRALI was induced by MHC-I antibodies. In the intervention group, C1-inhibitor was infused concomitantly. Mice were supported with mechanical ventilation. After 2 h, mice were killed, lungs were removed and bronchoalveolar lavage fluid (BALF) was obtained. RESULTS: Injection of MHC-I antibodies induced TRALI, illustrated by an increase in wet-to-dry ratio of the lungs, in BALF protein levels and in lung injury scores. TRALI was further characterized by complement activation, demonstrated by increased BALF levels of C3a and C5a. Administration of C1-inhibitor resulted in increased pulmonary C1-inhibitor levels with high activity. C1-inhibitor reduced pulmonary levels of complement C3a associated with improved lung injury scores. However, levels of pro-inflammatory mediators were unaffected. CONCLUSION: In a murine model of TRALI, C1-inhibitor attenuated pulmonary levels of C3a associated with improved lung injury scores, but with persistent high levels of inflammatory cytokines.


Asunto(s)
Lesión Pulmonar Aguda/tratamiento farmacológico , Proteína Inhibidora del Complemento C1/administración & dosificación , Reacción a la Transfusión , Reacción a la Transfusión/tratamiento farmacológico , Lesión Pulmonar Aguda/etiología , Lesión Pulmonar Aguda/patología , Análisis de Varianza , Animales , Anticuerpos/inmunología , Líquido del Lavado Bronquioalveolar/inmunología , Activación de Complemento/inmunología , Complemento C3a/inmunología , Complemento C5a/inmunología , Citocinas/inmunología , Modelos Animales de Enfermedad , Lipopolisacáridos , Pulmón/metabolismo , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Reacción a la Transfusión/patología
13.
Thromb Haemost ; 110(3): 582-92, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23783078

RESUMEN

Streptococcus pneumoniae is a common causative pathogen of pneumonia and sepsis. Pneumonia and sepsis are associated with enhanced activation of coagulation, resulting in the production of several host-derived proteases at the primary site of infection and in the circulation. Serine proteases cleave protease activated receptors (PARs), which form a molecular link between coagulation and inflammation. PAR4 is one of four subtypes of PARs and is widely expressed by multiple cell types in the respiratory tract implicated in pulmonary inflammation, by immune cells and by platelets. In mice, mouse (m)PAR4 is the only thrombin receptor expressed by platelets. We here sought to determine the contribution of mPAR4 to the host response during pneumococcal pneumonia. Pneumonia was induced by intranasal inoculation with S. pneumoniae in mPAR4-deficient (par4-/-) and wild-type mice. Mice were sacrificed after 6, 24 or 48 hours (h). Blood, lungs, liver and spleen were collected for analyses. Ex vivo stimulation assays were performed with S. pneumoniae and mPAR4 activating peptides. At 48 h after infection, higher bacterial loads were found in the lungs and blood of par4-/- mice (p < 0.05), accompanied by higher histopathology scores and increased cytokine levels (p < 0.05) in the lungs. Ex vivo, co-stimulation with mPAR4 activating peptide enhanced the whole blood cytokine response to S. pneumoniae. Thrombin inhibition resulted in decreased cytokine release after S. pneumoniae stimulation in human whole blood. Our findings suggest that mPAR4 contributes to antibacterial defence during murine pneumococcal pneumonia.


Asunto(s)
Pulmón/microbiología , Pulmón/patología , Neumonía Neumocócica/patología , Receptores de Trombina/metabolismo , Streptococcus pneumoniae/crecimiento & desarrollo , Animales , Plaquetas/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Inflamación , Hígado/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Péptidos/química , Neumonía Neumocócica/metabolismo , Neumonía Neumocócica/microbiología , Sepsis/metabolismo , Bazo/microbiología , Células Madre , Factores de Tiempo
14.
Crit Care Res Pract ; 2012: 720950, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22701787

RESUMEN

Purpose. Onset of transfusion-related acute lung injury (TRALI) is suggested to be a threshold-event. Data is lacking on the relation between titer of antibodies infused and onset of TRALI. We determined whether onset of TRALI is dependent on the titer of MHC-I antibodies infused in a combined model of ventilator-induced lung injury and antibody-induced TRALl. Methods. BALB/c mice were ventilated for five hours with low (7.5 ml/kg) or high (15 ml/kg) tidal volume. After three hours of MV, TRALI was induced by infusion of 0.5 mg/kg, 2.0 mg/kg or 4.5 mg/kg MHC-I antibodies. Control animals received vehicle. After five hours of MV, animals were sacrificed. Results. MV with high tidal volumes resulted in increased levels of all markers of lung injury compared to animals ventilated with low tidal MV. In ventilator-induced lung injury, infusion of 4.5 mg/kg of antibodies further increased pulmonary wet-to-dry ratio, pulmonary neutrophil influx and pulmonary KC levels, whereas infusion of lower dose of antibodies did not augment lung injury. In contrast, mice ventilated with low tidal volumes did not develop lung injury, irrespective of the dose of antibody used. Conclusions. In the presence of injurious MV, onset of TRALI depends on the titer of antibodies infused.

16.
J Thromb Haemost ; 9(10): 2020-8, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21848642

RESUMEN

BACKGROUND: Melioidosis is a frequent cause of sepsis in Southeast Asia caused by the Gram-negative bacterium Burkholderia pseudomallei. Patients with melioidosis have elevated circulating levels of plasminogen activator inhibitor type 1 (PAI-1), an important regulator of inflammation and fibrinolysis. OBJECTIVES: In this study, we aimed to investigate the role of PAI-1 during melioidosis. METHODS: Wild-type (WT) and PAI-1-deficient (PAI-1-/1(-/-) ) mice were intranasally infected with B. pseudomallei. Mice were killed after 24, 48 or 72 h. Lungs, liver and blood were harvested for measurement of bacterial loads, cytokines, clinical chemistry, histopathology, and coagulation parameters. Additionally, survival studies were performed. RESULTS: PAI-1(-/-) mice demonstrated enhanced susceptibility to B. pseudomallei infection, as shown by a strongly increased mortality rate (100% vs. 58% among WT mice, P < 0.001), associated with enhanced bacterial loads in lungs, liver, and blood. Additionally, PAI-1(-/-) mice showed elevated levels of proinflammatory cytokines in lungs and plasma, accompanied by enhanced local and systemic coagulation activation (thrombin-antithrombin complexes and D-dimer), increased hepatocellular injury (plasma aspartate aminotransferase and alanine aminotransferase), and renal failure (plasma creatinine and urea). CONCLUSIONS: PAI-1 has a protective role during severe Gram-negative sepsis caused by B. pseudomallei by limiting bacterial growth, inflammation, and coagulation, and probably, as a consequence thereof, distant organ injury.


Asunto(s)
Burkholderia pseudomallei/aislamiento & purificación , Melioidosis/inmunología , Inhibidor 1 de Activador Plasminogénico/sangre , Sepsis/inmunología , Animales , Burkholderia pseudomallei/crecimiento & desarrollo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Sepsis/microbiología
17.
J Thromb Haemost ; 9(1): 122-32, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21029363

RESUMEN

BACKGROUND: Streptococcus (S.) pneumoniae is the most common causative pathogen in community-acquired pneumonia and a major cause of sepsis. Recombinant human tissue factor pathway inhibitor (rh-TFPI) attenuates sepsis-induced coagulation and has been evaluated in clinical trials involving patients with sepsis and community-acquired pneumonia. OBJECTIVE: To examine the effect of rh-TFPI on coagulation, inflammation and bacterial outgrowth in S. pneumoniae pneumonia in mice, with or without concurrent antibiotic treatment. METHODS: Pneumonia was induced by intranasal inoculation with S. pneumoniae. Mice were treated with placebo, rh-TFPI, ceftriaxone or rh-TFPI combined with ceftriaxone. Early (8 h) and late (24 h) initiated treatments were evaluated. Samples were obtained 24 or 48 h after infection, for early and late initiated treatment, respectively. In vitro, placebo or rh-TFPI was added to a suspension of S. pneumoniae. RESULTS: Rh-TFPI reduced pneumonia-induced coagulation; rh-TFPI with ceftriaxone further attenuated coagulation relative to ceftriaxone alone. Rh-TFPI inhibited accumulation of neutrophils in lung tissue and reduced the levels of several cytokines and chemokines in lungs and plasma in mice not treated with antibiotics; in these animals, rh-TFPI initiated 24 h after infection decreased pulmonary bacterial loads. In vitro, rh-TFPI also inhibited growth of S. pneumoniae. CONCLUSIONS: Therapeutic rh-TFPI attenuates coagulation, inflammation and bacterial growth during pneumococcal pneumonia, whereby the latter two effects only become apparent in the absence of concurrent antibiotic treatment.


Asunto(s)
Antiinfecciosos/farmacología , Antiinflamatorios/farmacología , Anticoagulantes/farmacología , Lipoproteínas/farmacología , Neumonía Neumocócica/tratamiento farmacológico , Animales , Antiinfecciosos/administración & dosificación , Antiinfecciosos/farmacocinética , Antiinflamatorios/administración & dosificación , Antiinflamatorios/farmacocinética , Anticoagulantes/administración & dosificación , Anticoagulantes/farmacocinética , Coagulación Sanguínea/efectos de los fármacos , Ceftriaxona/farmacología , Modelos Animales de Enfermedad , Quimioterapia Combinada , Femenino , Humanos , Mediadores de Inflamación/metabolismo , Inyecciones Intraperitoneales , Lipoproteínas/administración & dosificación , Lipoproteínas/farmacocinética , Ratones , Ratones Endogámicos C57BL , Neumonía Neumocócica/sangre , Neumonía Neumocócica/inmunología , Neumonía Neumocócica/microbiología , Proteínas Recombinantes/farmacología , Streptococcus pneumoniae/efectos de los fármacos , Streptococcus pneumoniae/crecimiento & desarrollo , Factores de Tiempo
18.
Eur Respir J ; 36(6): 1346-54, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20413539

RESUMEN

Influenza A is a major cause of mortality. Knowledge on coagulation activation in influenza infection is limited. The factor V Leiden (FVL) mutation is possibly subject to positive selection pressure. It is unknown whether this mutation impacts on the outcome of severe influenza. In the present study, the effect of lethal influenza on pulmonary and systemic coagulation activation and whether or not FVL mutation alters coagulation activation in and the course of lethal influenza, was determined. Wild-type mice, and mice heterozygous or homozygous for FVL were infected intranasally with a lethal dose of H1N1 (haemagglutinin 1 and neuraminidase 1) influenza A. Mice were sacrificed after 48 or 96 h for determination of coagulation activation, histopathology, pulmonary inflammatory parameters and viral load, or were observed in a survival study. Extensive local and systemic coagulation activation during lethal influenza was demonstrated by increased lung and plasma levels of thrombin-antithrombin complexes and fibrin degradation products, and by pulmonary fibrin deposition. FVL mutation did not influence the procoagulant response, lung histopathology or survival. FVL mice demonstrated elevated viral loads 48 h after infection. In conclusion, coagulation is activated locally and systemically during lethal murine influenza A infection. The FVL mutation does not influence coagulation activation, lung inflammation or survival in lethal influenza A.


Asunto(s)
Trastornos de la Coagulación Sanguínea/genética , Factor V/genética , Subtipo H1N1 del Virus de la Influenza A , Infecciones por Orthomyxoviridae/sangre , Infecciones por Orthomyxoviridae/mortalidad , Animales , Antitrombinas/análisis , Factores de Coagulación Sanguínea/análisis , Femenino , Productos de Degradación de Fibrina-Fibrinógeno/análisis , Heterocigoto , Homocigoto , Pulmón/metabolismo , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación Puntual , Índice de Severidad de la Enfermedad , Trombina/análisis , Carga Viral
19.
Intensive Care Med ; 36(5): 879-87, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20221752

RESUMEN

PURPOSE: Transfusion-related acute lung injury (TRALI) occurs more often in critically ill patients than in a general hospital population, possibly due to the presence of underlying inflammatory conditions that may prime pulmonary neutrophils. Mechanical ventilation may be a risk factor for developing TRALI. We examined the influence of mechanical ventilation (MV) on the development of TRALI, combining a murine MV model causing ventilator-induced lung injury with a model of antibody-induced TRALl. METHODS: BALB/c mice (n = 84) were ventilated for 5 h with low (7.5 ml/kg) or high (15 ml/kg) tidal volume, a positive end-expiratory pressure of 2 cm H(2)O and a fraction of inspired oxygen of 50%. After 3 h of MV, TRALI was induced by infusion of MHC-I antibodies (4.5 mg/kg); controls received vehicle. Non-ventilated animals receiving vehicle, isotype or MHC-I antibodies served as additional controls. RESULTS: All animals receiving MHC-I antibodies developed TRALI within 2 h. In mice in which TRALI was induced, MV with low tidal volumes aggravated pulmonary injury, as evidenced by an increase in neutrophil influx, pulmonary and systemic levels of cytokines and lung histopathological changes compared to unventilated controls. The use of high tidal volume ventilation resulted in a further increase in protein leakage and pulmonary edema. CONCLUSIONS: Mechanical ventilation (MV) synergistically augmented lung injury during TRALI, which was even further enhanced by the use of injurious ventilator settings. Results suggest that MV may be a risk factor for the onset of TRALI and may aggravate the course of disease.


Asunto(s)
Lesión Pulmonar Aguda/etiología , Antígenos de Histocompatibilidad Clase II/inmunología , Neutrófilos/inmunología , Reacción a la Transfusión , Lesión Pulmonar Inducida por Ventilación Mecánica/inmunología , Ventiladores Mecánicos/efectos adversos , Lesión Pulmonar Aguda/inmunología , Animales , Anticuerpos/inmunología , Análisis de los Gases de la Sangre , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/inmunología , Enfermedad Crítica , Modelos Animales de Enfermedad , Leucocitos/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Factores de Riesgo
20.
J Infect Dis ; 198(8): 1189-97, 2008 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-18752445

RESUMEN

BACKGROUND: Ixodes ticks are the main vectors for Borrelia burgdorferi sensu lato. In the United States, B. burgdorferi is the sole causative agent of Lyme borreliosis and is transmitted by Ixodes scapularis. In Europe, 3 Borrelia species-B. burgdorferi, B. garinii, and B. afzelii-are prevalent, which are transmitted by Ixodes ricinus. The I. scapularis salivary protein Salp15 has been shown to bind to B. burgdorferi outer surface protein (Osp) C, protecting the spirochete from antibody-mediated killing. METHODS AND RESULTS: We recently identified a Salp15 homologue in I. ricinus, Salp15 Iric-1. Here, we have demonstrated, by solid-phase overlays, enzyme-linked immunosorbent assay, and surface plasmon resonance, that Salp15 Iric-1 binds to B. burgdorferi OspC. Importantly, this binding protected the spirochete from antibody-mediated killing in vitro and in vivo; immune mice rechallenged with B. burgdorferi preincubated with Salp15 Iric-1 displayed significantly higher Borrelia numbers and more severe carditis, compared with control mice. Furthermore, Salp15 Iric-1 was capable of binding to OspC from B. garinii and B. afzelii, but these Borrelia species were not protected from antibody-mediated killing. CONCLUSIONS: Salp15 Iric-1 interacts with all European Borrelia species but differentially protects B. burgdorferi from antibody-mediated killing, putatively giving this Borrelia species a survival advantage in nature.


Asunto(s)
Antígenos Bacterianos/metabolismo , Proteínas de la Membrana Bacteriana Externa/metabolismo , Borrelia burgdorferi/efectos de los fármacos , Borrelia burgdorferi/metabolismo , Ixodes/microbiología , Saliva/metabolismo , Proteínas y Péptidos Salivales/farmacología , Animales , Anticuerpos Antibacterianos/sangre , Anticuerpos Antibacterianos/inmunología , Borrelia burgdorferi/inmunología , Borrelia burgdorferi/patogenicidad , Ensayo de Inmunoadsorción Enzimática , Femenino , Ratones , Ratones Endogámicos C3H , Conejos , Proteínas y Péptidos Salivales/metabolismo , Resonancia por Plasmón de Superficie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...