Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Autoimmun ; 65: 19-29, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26297208

RESUMEN

In viral infection, morbidity and mortality often result from extrahepatic disease manifestations such as vasculitis. We hereby show that human microvascular endothelial cells express viral receptors of the innate immune system which are induced upon ligand engagement. Furthermore, stimulation of endothelial cells with the synthetic analog of viral DNA, poly (dA:dT), human DNA and hepatitis B virus-containing immunoprecipitates from a patient with polyarteritis nodosa induces an inflammatory response including the upregulation of adhesion molecules, which is mediated exclusively by TLR9 and involves an IRF3-dependent pathway. Thus, endothelial cells are able to actively participate in immune mediated vascular inflammation caused by viral infections. Furthermore, we provide evidence for the ability of LL37 to bind and internalize viral or endogenous DNA into non-immune cells. DNA nucleotides internalized by LL37 suppress the production of proinflammatory mediators suggesting a protective effect against direct responses to viral infection or circulating DNA-fragments of endogenous origin.


Asunto(s)
Catelicidinas/inmunología , ADN Viral/inmunología , Células Endoteliales/inmunología , Inflamación/inmunología , Microvasos/inmunología , Poli dA-dT/inmunología , Péptidos Catiónicos Antimicrobianos , Catelicidinas/metabolismo , Células Cultivadas , Quimiocinas/inmunología , Quimiocinas/metabolismo , Trampas Extracelulares/metabolismo , Virus de la Hepatitis B/inmunología , Células Endoteliales de la Vena Umbilical Humana/inmunología , Humanos , Inflamación/metabolismo , Inflamación/virología , Factor 3 Regulador del Interferón/inmunología , Transducción de Señal/inmunología , Receptor Toll-Like 9/inmunología
2.
Arterioscler Thromb Vasc Biol ; 32(12): 2884-91, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23023375

RESUMEN

OBJECTIVE: Hydrogen sulfide (H(2)S)-releasing NSAIDs exert potent anti-inflammatory effects beyond classical cyclooxygenase inhibition. Here, we compared the platelet inhibitory effects of the H(2)S-releasing aspirin derivative ACS14 with its mother compound aspirin to analyze additional effects on platelets. METHODS AND RESULTS: In platelets of mice fed with ACS14 for 6 days (50 mg/kg per day), not only arachidonic acid-induced platelet aggregation but also ADP-dependent aggregation was decreased, an effect that was not observed with an equimolar dose of aspirin (23 mg/kg per day). ACS14 led to a significantly longer arterial occlusion time after light-dye-induced endothelial injury as well as decreased thrombus formation after ferric chloride-induced injury in the carotid artery. Bleeding time was not prolonged compared with animals treated with equimolar doses of aspirin. In vitro, in human whole blood, ACS14 (25-500 µmol/L) inhibited arachidonic acid-induced platelet aggregation, but compared with aspirin additionally reduced thrombin receptor-activating peptide-, ADP-, and collagen-dependent aggregation. In washed human platelets, ACS14 (500 µmol/L) attenuated αIIbß3 integrin activation and fibrinogen binding and increased intracellular cAMP levels and cAMP-dependent vasodilator-stimulated phosphoprotein (VASP) phosphorylation. CONCLUSIONS: The H(2)S-releasing aspirin derivative ACS14 exerts strong antiaggregatory effects by impairing the activation of the fibrinogen receptor by mechanisms involving increased intracellular cyclic nucleotides. These additional antithrombotic properties result in a more efficient inhibition of thrombus formation in vivo as achieved with aspirin alone.


Asunto(s)
Aspirina/metabolismo , Aspirina/farmacología , Plaquetas/efectos de los fármacos , Fibrinolíticos/farmacología , Sulfuro de Hidrógeno/metabolismo , Agregación Plaquetaria/efectos de los fármacos , Animales , Aspirina/análogos & derivados , Tiempo de Sangría , Plaquetas/metabolismo , AMP Cíclico/metabolismo , Disulfuros/farmacología , Humanos , Técnicas In Vitro , Integrinas/efectos de los fármacos , Integrinas/metabolismo , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Activación Plaquetaria/efectos de los fármacos , Activación Plaquetaria/fisiología , Agregación Plaquetaria/fisiología , Prostaglandina-Endoperóxido Sintasas/efectos de los fármacos , Prostaglandina-Endoperóxido Sintasas/metabolismo , Trombosis/metabolismo , Trombosis/prevención & control
3.
Mediators Inflamm ; 2013: 279781, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23766558

RESUMEN

INTRODUCTION: Inflammation and endothelium-derived superoxides are important pathomechanisms in atherothrombotic diseases. We could previously show that the tyrosine phosphatase SHP-1 acts as a negative regulator in endothelial superoxide production. In this study we investigated the influence of SHP-1 on platelet-endothelium interaction and arterial thrombosis in TNFα -induced endothelial inflammation in vivo. METHODS: Arteriolar thrombosis and platelet rolling in vivo were investigated in C57BL/6 mice using intravital microscopy in the dorsal skinfold chamber microcirculation model. RESULTS: Inhibition of SHP-1 by the specific pharmacological inhibitor sodium stibogluconate did not significantly enhance platelet-endothelium interaction in vivo under physiological conditions but led to an augmented fraction of rolling platelets in TNFα -induced systemic inflammation. Accordingly, ferric-chloride-induced arteriolar thrombus formation, which was already increased by SHP-1 inhibition, was further enhanced in the setting of TNFα -induced inflammation. Platelet aggregation in vitro as well as ex vivo was not influenced by SHP-1-inhibition. In cultured endothelial cells, sodium stibogluconate increased TNFα -induced surface expression of p-selectin and von Willebrand factor. Additionally, TNFα increased SHP-1 activity and protein expression. CONCLUSIONS: The endothelial tyrosine phosphatase SHP-1 plays an important role for vascular hemostasis in vivo, which is crucial in TNF α -induced endothelial inflammation where it may serve as an autoinhibitory molecule to prevent excess inflammatory response and thrombus formation.


Asunto(s)
Plaquetas/metabolismo , Endotelio/metabolismo , Inflamación/inducido químicamente , Inflamación/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Animales , Gluconato de Sodio Antimonio/farmacología , Western Blotting , Células Cultivadas , Humanos , Ratones , Ratones Endogámicos C57BL , Proteína Tirosina Fosfatasa no Receptora Tipo 6/antagonistas & inhibidores , Porcinos
4.
Mol Cell Biochem ; 370(1-2): 151-61, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22864531

RESUMEN

Vascular endothelial growth factor (VEGF) plays an important role in glomerular homeostasis as well as in the pathogenesis of kidney diseases as glomerulonephritis (GN) and diabetic nephropathy. Mesangial cells (MC), which are an integral part of the functional glomerular filtration barrier in that providing structural support, can behave like inflammatory cells and produce mediators as chemokines and growth factors; they are known to express viral receptors, with TLR3 having been attributed relevance in viral disease-associated GN. Experiments were performed on human MC in cell culture. Stimulation experiments were performed with poly (I:C) and hepatitis C RNA from patients with hepatitis C infection. We hereby show a TLR3-mediated upregulation of VEGF and its receptor subtype 2 (VEGF-R2) in human MC upon activation of viral receptors by poly (I:C) and hepatitis C virus. The increase in VEGF expression levels is further enhanced by tumor necrosis factor alpha (TNFα) which also induces the cytokines IL-6 and IL-8 as well as the chemokines MCP-1 and RANTES. These effects are potentiated by preincubation of MC with poly (I:C), just as the induction of the viral receptors TLR3, RIG-1, and MDA5 themselves. Moreover, MCP-1 itself is able to significantly increase mesangial VEGF expression. Therefore, with VEGF and VEGF-R2 being induced upon viral receptor activation in human MC, a novel role of TLR3 in mediating glomerular damage in virally induced or aggravated GN is inferred. TNFα and MCP-1 are seemingly important in amplifying VEGF effects in the setting of virally induced inflammation, with TNFα being also able to induce other mediators of glomerular pathology in GN.


Asunto(s)
Células Mesangiales/metabolismo , Receptores Virales/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Quimiocinas/metabolismo , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Hepacivirus/efectos de los fármacos , Hepacivirus/fisiología , Humanos , Mediadores de Inflamación/farmacología , Helicasa Inducida por Interferón IFIH1 , Células Mesangiales/efectos de los fármacos , Células Mesangiales/virología , Poli I-C/farmacología , ARN Interferente Pequeño/metabolismo , Receptores de Ácido Retinoico/genética , Receptores de Ácido Retinoico/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular/genética , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor Toll-Like 3/genética , Receptor Toll-Like 3/metabolismo , Transfección , Factor A de Crecimiento Endotelial Vascular/genética
5.
Pharm Res ; 29(5): 1282-94, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22274557

RESUMEN

PURPOSE: Site specific vascular gene delivery is a promising tool for treatment of cardiovascular diseases. By combining ultrasound mediated microbubble destruction with site specific magnetic targeting of lentiviruses, we aimed to develop a technique suitable for systemic application. METHODS: The magnetic nanoparticle coupling to lipid microbubbles was confirmed by absorbance measurements. Association of fluorescent lentivirus to magnetic microbubbles (MMB) was determined by microscopy and flow cytometry. Functionality and efficiency of GFP-encoding lentiviral MMB transduction was evaluated by endothelial (HMEC) GFP expression and cytotoxicity was measured by MTT reduction. RESULTS: Microbubbles with a mean diameter of 4.3 ± 0.04 µm were stable for 2 days, readily magnetizable and magnetically steerable in vitro and efficiently associated with lentivirus. Exposure of eGFP-encoding lentiviral MMB to human endothelial cells followed by application of an external static magnetic field (30 min) and ultrasonic destruction of the microbubbles did not markedly affect cellular viability. Finally, this combination led to a 30-fold increase in transduction efficiency compared to application of naked virus alone. CONCLUSIONS: By associating microbubbles with magnetic iron nanoparticles, these function as carriers for lentiviruses achieving tissue specific deposition at the site of interest.


Asunto(s)
Células Endoteliales/metabolismo , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Lentivirus/genética , Magnetismo , Microburbujas , Ultrasonido , Supervivencia Celular , Sistemas de Liberación de Medicamentos , Células Endoteliales/citología , Citometría de Flujo , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Tamaño de la Partícula
6.
Nanomedicine ; 8(8): 1309-18, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22480917

RESUMEN

Site specific vascular gene delivery for therapeutic implications is favorable because of reduction of possible side effects. Yet this technology faces numerous hurdles that result in low transfection rates because of suboptimal delivery. Combining ultrasonic microbubble technology with magnetic nanoparticle enhanced gene transfer could make it possible to use the systemic vasculature as the route of application and to magnetically trap these compounds at the target of interest. In this study we show that magnetic nanoparticle-coated microbubbles bind plasmid DNA and successfully deliver it to endothelial cells in vitro and more importantly transport their cargo through the vascular system and specifically deliver it to the vascular wall in vivo at sites where microbubbles are retained by magnetic force and burst by local ultrasound application. This resulted in a significant enhancement in site specific gene delivery compared with the conventional microbubble technique. Thus, this technology may have promising therapeutic potential. FROM THE CLINICAL EDITOR: This work focuses on combining ultrasonic microbubble technology with magnetic nanoparticle enhanced gene transfer to enable targeted gene delivery via the systemic vasculature and magnetic trapping of these compounds at the target of interest.


Asunto(s)
Sistemas de Liberación de Medicamentos , Técnicas de Transferencia de Gen , Nanopartículas de Magnetita , Microburbujas , Células Endoteliales , Terapia Genética , Humanos , Nanopartículas de Magnetita/administración & dosificación , Nanopartículas de Magnetita/química , Plásmidos , Ultrasonido
7.
Eur Heart J ; 32(4): 476-84, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21036775

RESUMEN

AIMS: We sought to determine whether circulating vascular progenitor cells, such as endothelial progenitor cells (EPCs) or smooth muscle progenitor cells (SPCs), were associated with the severity of cardiac allograft vasculopathy (CAV). METHODS AND RESULTS: CD34(+)CD140b(+) SPCs and CD34(+)KDR(+) EPCs were measured in the peripheral circulation of 187 adult heart transplant recipients by flow cytometry. Cardiac allograft vasculopathy was quantified by angiography using a CAV-specific scoring system. Cardiac allograft vasculopathy was present in 84 patients (44.7%) and was classified as mild in 59 and severe in 25 cases. Circulating SPCs were more frequently detectable in CAV patients than in patients without CAV. The number of CD34(+)CD140b(+) cells showed a stepwise increase in patients with moderate and severe CAV. Smooth muscle progenitor cell counts were higher in patients with coronary stent implant compared with unstented patients with CAV. In contrast, peripheral CD34(+)KDR(+) EPC counts were not changed in CAV patients. Plasma CXCL12 levels correlated with the degree of CAV and SPC counts. None of the different immunosuppressive drug regimes was related to the SPC count or the CXCL12 levels. A multivariate regression analysis revealed that the SPC count was independently associated with the presence of CAV. CONCLUSION: Circulating SPCs, but not EPCs, and plasma CXCL12 concentrations are elevated in CAV patients, indicating that they play prominent roles in transplant arteriosclerosis.


Asunto(s)
Antígenos CD34/metabolismo , Quimiocina CXCL12/metabolismo , Rechazo de Injerto/etiología , Trasplante de Corazón/patología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Células Madre/patología , Adulto , Anciano , Proliferación Celular , Células Endoteliales/patología , Endotelio Vascular/patología , Femenino , Citometría de Flujo , Rechazo de Injerto/patología , Humanos , Masculino , Persona de Mediana Edad , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Trasplante Homólogo
8.
Cytometry A ; 75(10): 848-53, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19739088

RESUMEN

Circulating adult CD34(+)VEGFR2(+) endothelial progenitor cells (EPCs) have been shown to differentiate into endothelial cells, thus contributing to vascular homeostasis. Furthermore, a subset of circulating CD14(+) monocytes coexpresses CD16 together with the angiopoietin receptor Tie2 and has been functionally implicated in tumor angiogenesis. However, clinically applicable protocols for flow cytometric quantification of EPCs and Tie2(+) monocytes in peripheral blood and a consensus on reference values remain elusive. The number of Tie2(+)CD14(+)CD16(mid) angiogenic monocytes and CD34(+)VEGFR2(+)CD45(low/-) EPCs was assessed in the peripheral venous blood of patients with stable coronary artery disease by three-color flow cytometry using specific monoclonal antibodies conjugated to PerCP, PE, PE-Cy7, APC, and APC-Cy7. Scatter multigating with exclusion of dead cells was performed to dissect complex mononuclear cell populations. This analysis was further refined by matching bright fluorochromes (PE-Cy7, PE, APC) with dimly expressed markers (CD34, VEGFR2, Tie2), by automatic compensation for minimizing fluorescence spillover and by using fluorescence-minus-one (FMO) controls to determine positive/negative boundaries. Presuming a Gaussian distribution, we obtained average values (mean +/- SD) of 1.45 +/- 1.29% for Tie2(+)CD14(+)CD16(mid) monocytes (n = 11, range: 0.12-3.64%) and 0.019 +/- 0.013% for CD34(+)VEGFR2(+)CD45(low/-) EPCs (n = 17, range: 0.003-0.042%). The intra- and inter-assay variability was 1.6% and 4.5%, respectively. We have optimized a fast and sensitive assay for the flow cytometric quantification of circulating angiogenic monocytes and EPCs in cardiovascular medicine. This protocol may represent a basis for standardized analysis and monitoring of these cell subsets to define their normal range and prognostic/diagnostic value in clinical use.


Asunto(s)
Células Endoteliales/citología , Citometría de Flujo/métodos , Monocitos/citología , Neovascularización Fisiológica , Células Madre/citología , Anciano , Células Endoteliales/metabolismo , Femenino , Humanos , Antígenos Comunes de Leucocito/metabolismo , Receptores de Lipopolisacáridos/metabolismo , Masculino , Receptor TIE-2/metabolismo , Receptores de IgG/metabolismo , Células Madre/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
9.
Mol Cell Biochem ; 322(1-2): 193-206, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19005739

RESUMEN

Viral inflammation and infection of mesothelial cells (MC) are a major problem in several organ systems including pleura, pericardium and peritoneum. Toll-like receptors (TLRs) are an essential part of the innate immune system for early recognition of pathogen-associated molecular patterns. TLRs recognise molecular patterns associated with microbial pathogens and induce an immune response. TLR3 recognises dsRNA of viral origin as exemplified by poly (I:C) RNA, a synthetic analogue of viral dsRNA. The helicases RIG-I and MDA5 may also act as sensors of viral infections. MC exhibit an expression of TLR3, RIG-I and MDA5. Poly (I:C) RNA stimulation resulted in an up-regulation of proinflammatory cytokines and chemokines as well as type I interferons. This novel finding of functional expression of viral sensors on human MC may indicate a novel link between viral infections and mesothelial inflammation and indicates a pathophysiologic role of viral receptors in these processes.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , Células Epiteliales/metabolismo , Mediadores de Inflamación/metabolismo , Poli I-C/farmacología , Receptor Toll-Like 3/metabolismo , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/antagonistas & inhibidores , ARN Helicasas DEAD-box/genética , Humanos , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Helicasa Inducida por Interferón IFIH1 , ARN Mensajero/metabolismo , ARN Viral/metabolismo , Receptores Inmunológicos , Receptor Toll-Like 3/antagonistas & inhibidores , Receptor Toll-Like 3/genética , Transfección
10.
EBioMedicine ; 42: 120-132, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30905847

RESUMEN

BACKGROUND: Sepsis, the most severe form of infection, involves endothelial dysfunction which contributes to organ failure. To improve therapeutic prospects, elucidation of molecular mechanisms underlying endothelial vascular failure is of essence. METHODS: Polymicrobial contamination induced sepsis mouse model and primary endothelial cells incubated with sepsis serum were used to study SHP-2 in sepsis-induced endothelial inflammation. SHP-2 activity was assessed by dephosphorylation of pNPP, ROS production was measured by DCF oxidation and protein interactions were assessed by proximity ligation assay. Vascular inflammation was studied in the mouse cremaster model and in an in vitro flow assay. FINDINGS: We identified ROS-dependent inactivation of the tyrosine phosphatase SHP-2 to be decisive for endothelial activation in sepsis. Using in vivo and in vitro sepsis models, we observed a significant reduction of endothelial SHP-2 activity, accompanied by enhanced adhesion molecule expression. The impaired SHP-2 activity was restored by ROS inhibitors and an IL-1 receptor antagonist. SHP-2 activity inversely correlated with the adhesive phenotype of endothelial cells exposed to IL-1ß as well as sepsis serum via p38 MAPK and NF-κB. In vivo, SHP-2 inhibition accelerated IL-1ß-induced leukocyte adhesion, extravasation and vascular permeability. Mechanistically, SHP-2 directly interacts with the IL-1R1 adaptor protein MyD88 via its tyrosine 257, resulting in reduced binding of p85/PI3-K to MyD88. INTERPRETATION: Our data show that SHP-2 inactivation by ROS in sepsis releases a protective break, resulting in endothelial activation. FUND: German Research Foundation, LMU Mentoring excellence and FöFoLe Programme, Verein zur Förderung von Wissenschaft und Forschung, German Ministry of Education and Research.


Asunto(s)
Endotelio Vascular/metabolismo , Endotelio Vascular/fisiopatología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Sepsis/metabolismo , Sepsis/fisiopatología , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Activación Enzimática , Femenino , Humanos , Mediadores de Inflamación/metabolismo , Leucocitos/metabolismo , Masculino , Ratones , FN-kappa B/metabolismo , Fosforilación , Unión Proteica , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Especies Reactivas de Oxígeno/metabolismo , Sepsis/etiología
11.
J Vasc Res ; 45(2): 153-63, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-17962719

RESUMEN

Endothelial cell survival is indispensable to maintain endothelial integrity and initiate new vessel formation. We investigated the role of SHP-2 in endothelial cell survival and angiogenesis in vitro as well as in vivo. SHP-2 function in cultured human umbilical vein and human dermal microvascular endothelial cells was inhibited by either silencing the protein expression with antisense-oligodesoxynucleotides or treatment with a pharmacological inhibitor (PtpI IV). SHP-2 inhibition impaired capillary-like structure formation (p < 0.01; n = 8) in vitro as well as new vessel growth ex vivo(p < 0.05; n = 10) and in vivo in the chicken chorioallantoic membrane (p < 0.01, n = 4). Additionally, SHP-2 knock-down abrogated fibroblast growth factor 2 (FGF-2)-dependent endothelial proliferation measured by MTT reduction (p < 0.01; n = 12). The inhibitory effect of SHP-2 knock-down on vessel growth was mediated by increased endothelial apoptosis (annexin V staining, p < 0.05, n = 9), which was associated with reduced FGF-2-induced phosphorylation of phosphatidylinositol 3-kinase (PI3-K), Akt and extracellular regulated kinase 1/2 (ERK1/2) and involved diminished ERK1/2 phosphorylation after PI3-K inhibition (n = 3). These results suggest that SHP-2 regulates endothelial cell survival through PI3-K-Akt and mitogen-activated protein kinase pathways thereby strongly affecting new vessel formation. Thus, SHP-2 exhibits a pivotal role in angiogenesis and may represent an interesting target for therapeutic approaches controlling vessel growth.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Membrana Corioalantoides/irrigación sanguínea , Células Endoteliales/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Oligonucleótidos Antisentido/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Embrión de Pollo , Células Endoteliales/enzimología , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Humanos , Ratones , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Técnicas de Cultivo de Tejidos , Transfección
12.
Arterioscler Thromb Vasc Biol ; 27(12): 2540-7, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17932320

RESUMEN

OBJECTIVE: Hypoxia-inducible factor (HIF)-1alpha is the regulatory subunit of a transcriptional complex, which controls the recruitment of multipotent progenitor cells and tissue repair in ischemic tissue by inducing stromal cell-derived factor (SDF)-1alpha expression. Because HIF-1alpha can be activated under normoxic conditions in smooth muscle cells (SMCs) by platelet products, we investigated the role of HIF-1alpha in SDF-1alpha-mediated neointima formation after vascular injury. METHODS AND RESULTS: Wire-induced injury of the left carotid artery was performed in apolipoprotein E-deficient mice. HIF-1alpha expression was increased in the media as early as 1 day after injury, predominantly in SMCs. Nuclear translocation of HIF-1alpha and colocalization with SDF-1alpha was detected in neointimal cells after 2 weeks. HIF-1alpha mRNA expression was induced at 6 hours after injury as determined by real-time RT-PCR. Inhibition of HIF-1alpha expression by local application of HIF-1alpha-siRNA reduced the neointimal area by 49% and significantly decreased the neointimal SMCs content compared with control-siRNA. HIF-1alpha and SDF-1alpha expression were clearly diminished in neointimal cells of HIF-1alpha-siRNA treated arteries. CONCLUSIONS: HIF-1alpha expression is directly involved in neointimal formation after vascular injury and mediates the upregulation of SDF-1alpha, which may affect the stem cell-based repair of injured arteries.


Asunto(s)
Apolipoproteínas E/metabolismo , Traumatismos de las Arterias Carótidas/metabolismo , Arteria Carótida Común/metabolismo , Quimiocina CXCL12/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Túnica Íntima/metabolismo , Túnica Media/metabolismo , Transporte Activo de Núcleo Celular , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Traumatismos de las Arterias Carótidas/patología , Arteria Carótida Común/patología , Movimiento Celular , Modelos Animales de Enfermedad , Femenino , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocitos del Músculo Liso/metabolismo , Interferencia de ARN , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Células Madre/patología , Factores de Tiempo , Túnica Íntima/patología , Túnica Media/patología , Regulación hacia Arriba
13.
Vasc Health Risk Manag ; 4(3): 637-45, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18827913

RESUMEN

A common pathophysiological course in vascular diseases is an overwhelming activation and aggregation of blood platelets, which results in atherothrombosis. By causing the last decisive step of cerebral, coronary, or peripheral arterial ischemia thrombotic complications of atherosclerotic disease represent a major player in death cause statistics of most western countries. The development of novel therapies against platelet-dependent thrombosis and the concurrent improvement of existing therapeutic strategies thus is a paramount focus of pharmaceutical research. Currently, efficiency, dosing and indications of established antiplatelet substances are being re-evaluated, whilst new, so far unrecognized molecular targets for inhibition of platelet activity come up front. This not only allows for interesting new therapeutical options, but also widens our insight into the role platelets play in atherosclerosis in general. This article summarizes the relevant pathophysiology of platelet activation, presents current concepts in antiplatelet drug therapy, and highlights the role of platelets in vascular diseases apart from atherothrombosis.


Asunto(s)
Aterosclerosis/tratamiento farmacológico , Inhibidores de Agregación Plaquetaria/farmacología , Inhibidores de Agregación Plaquetaria/uso terapéutico , Trombosis/tratamiento farmacológico , Aterosclerosis/fisiopatología , Clopidogrel , Stents Liberadores de Fármacos , Humanos , Piperazinas/administración & dosificación , Activación Plaquetaria/fisiología , Agregación Plaquetaria/fisiología , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/antagonistas & inhibidores , Clorhidrato de Prasugrel , Antagonistas del Receptor Purinérgico P2 , Piridinas/uso terapéutico , Receptores de Tromboxanos/efectos de los fármacos , Transducción de Señal/fisiología , Tiofenos/administración & dosificación , Trombosis/fisiopatología , Ticlopidina/administración & dosificación , Ticlopidina/análogos & derivados , Ticlopidina/uso terapéutico
14.
J Clin Invest ; 111(11): 1665-72, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12782669

RESUMEN

Antimicrobial peptides are effector molecules of the innate immune system and contribute to host defense and regulation of inflammation. The human cathelicidin antimicrobial peptide LL-37/hCAP-18 is expressed in leukocytes and epithelial cells and secreted into wound and airway surface fluid. Here we show that LL-37 induces angiogenesis mediated by formyl peptide receptor-like 1 expressed on endothelial cells. Application of LL-37 resulted in neovascularization in the chorioallantoic membrane assay and in a rabbit model of hind-limb ischemia. The peptide directly activates endothelial cells, resulting in increased proliferation and formation of vessel-like structures in cultivated endothelial cells. Decreased vascularization during wound repair in mice deficient for CRAMP, the murine homologue of LL-37/hCAP-18, shows that cathelicidin-mediated angiogenesis is important for cutaneous wound neovascularization in vivo. Taken together, these findings demonstrate that LL-37/hCAP-18 is a multifunctional antimicrobial peptide with a central role in innate immunity by linking host defense and inflammation with angiogenesis and arteriogenesis.


Asunto(s)
Antibacterianos/química , Péptidos Catiónicos Antimicrobianos/farmacología , Neovascularización Fisiológica , Profármacos/farmacología , Receptores de Formil Péptido , Receptores de Lipoxina , Acetilcisteína/farmacología , Animales , Péptidos Catiónicos Antimicrobianos/genética , Apoptosis , Western Blotting , Calcio/metabolismo , Catelicidinas , División Celular , Células Cultivadas , Colágeno/farmacología , Cricetinae , Combinación de Medicamentos , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Inhibidores Enzimáticos/farmacología , Humanos , Indoles/farmacología , Inflamación/metabolismo , Isquemia , Laminina/farmacología , Maleimidas/farmacología , Mesocricetus , Ratones , FN-kappa B/metabolismo , Péptidos/química , Proteoglicanos/farmacología , Conejos , Receptores de Superficie Celular/biosíntesis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Cicatrización de Heridas
15.
Circ Res ; 96(1): 43-53, 2005 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-15569826

RESUMEN

The cardiovascular hormone atrial natriuretic peptide (ANP) exerts anti-inflammatory effects on tumor necrosis factor-alpha-activated endothelial cells by inducing mitogen-activated protein kinase (MAPK) phosphatase-1 (MKP-1). The underlying mechanisms are as yet unknown. We aimed to elucidate the signaling pathways leading to an induction of MKP-1 by ANP in primary human endothelial cells. By using antioxidants, generation of reactive oxygen species (ROS) was shown to be crucially involved in MKP-1 upregulation. ANP was found to increase ROS formation in cultured cells as well as in the endothelium of intact rat lung vessels. We applied NAD(P)H oxidase (Nox) inhibitors (apocynin and gp91ds-tat) and revealed this enzyme complex to be crucial for superoxide generation and MKP-1 expression. Moreover, by performing Nox2/4 antisense experiments, we identified Nox2 as the critically involved Nox homologue. Pull-down assays and confocal microscopy showed that ANP activates the small Rho-GTPase Rac1. Transfection of a dominant-negative (RacN17) and constitutively active Rac1 mutant (RacV12) indicated that ANP-induced superoxide generation and MKP-1 expression are mediated via Rac1 activation. ANP-evoked production of superoxide was found to activate c-Jun N-terminal kinase (JNK). Using specific inhibitors, we linked ANP-induced JNK activation to MKP-1 expression and excluded an involvement of protein kinase C, extracellular signal-regulated kinase, and p38 MAPK. MKP-1 induction was shown to depend on activation of the transcription factor activator protein-1 (AP-1) by using electrophoretic mobility shift assay and AP-1 decoys. In summary, our work provides insights into the mechanisms by which ANP induces MKP-1 and shows that ANP is a novel endogenous activator of endothelial Rac1 and Nox/Nox2.


Asunto(s)
Factor Natriurético Atrial/fisiología , Proteínas de Ciclo Celular/biosíntesis , GMP Cíclico/análogos & derivados , Células Endoteliales/efectos de los fármacos , Proteínas Inmediatas-Precoces/biosíntesis , Glicoproteínas de Membrana/fisiología , NADPH Oxidasas/fisiología , Fosfoproteínas Fosfatasas/biosíntesis , Proteínas Tirosina Fosfatasas/biosíntesis , Acetofenonas/farmacología , Animales , Factor Natriurético Atrial/farmacología , Capilares , Proteínas de Ciclo Celular/genética , Células Cultivadas/efectos de los fármacos , Células Cultivadas/metabolismo , GMP Cíclico/metabolismo , GMP Cíclico/farmacología , Cicloheximida/farmacología , ADN sin Sentido/farmacología , Fosfatasa 1 de Especificidad Dual , Células Endoteliales/metabolismo , Endotelio Vascular/citología , Inducción Enzimática/efectos de los fármacos , Inducción Enzimática/fisiología , Glicoproteínas/farmacología , Guanilato Ciclasa/efectos de los fármacos , Guanilato Ciclasa/fisiología , Humanos , Proteínas Inmediatas-Precoces/genética , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Pulmón/irrigación sanguínea , MAP Quinasa Quinasa 4 , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Glicoproteínas de Membrana/biosíntesis , Glicoproteínas de Membrana/genética , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , NADPH Oxidasa 1 , NADPH Oxidasa 2 , NADPH Oxidasa 4 , NADPH Oxidasa 5 , NADPH Oxidasas/biosíntesis , NADPH Oxidasas/genética , Oligonucleótidos Antisentido/farmacología , Fosfoproteínas Fosfatasas/genética , Proteína Fosfatasa 1 , Proteínas Tirosina Fosfatasas/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Receptores del Factor Natriurético Atrial/efectos de los fármacos , Receptores del Factor Natriurético Atrial/fisiología , Proteínas Recombinantes de Fusión/fisiología , Factor de Transcripción AP-1/metabolismo
16.
Arterioscler Thromb Vasc Biol ; 26(3): 508-13, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16373615

RESUMEN

OBJECTIVE: Previous studies have shown that elevated homocysteine (Hcy) levels promote the development of atherosclerotic lesions in atherosclerosis-prone animal models. There is evidence that oxidant stress contributes to Hcy's deleterious effects on the vasculature. The accumulation and adhesion of monocytes to the vascular endothelium is a critical event in the development of atherosclerosis. We investigated the effects of Hcy on the interaction between human endothelial cells (EC) (EC line EA.hy 926 and primary human umbilical vein EC [HUVEC]) and the monocytic cell line THP-1, and the impact of vascular oxidant stress and redox-sensitive signaling pathways on these events. METHODS AND RESULTS: L-Hcy, but not D-Hcy, increases the production of reactive oxygen species inside EC, enhances nuclear factor(NF)-kappaB activation, and stimulates intercellular adhesion molecule-1 (ICAM-1) RNA transcription and cell surface expression. This leads to a time- and dose-dependent increase in monocyte adhesion to ECs. Pretreatment of ECs with superoxide scavengers (MnTBAP and Tiron) or with an inhibitor of NF-kappaB activation abolished Hcy-induced monocyte adhesion, ICAM-1 expression, and nuclear translocation of NF-kappaB. CONCLUSIONS: These findings suggest that reactive oxygen species produced under hyperhomocysteinemic conditions may induce a proinflammatory situation in the vessel wall that initiates and promotes atherosclerotic lesion development.


Asunto(s)
Aterosclerosis/inmunología , Aterosclerosis/metabolismo , Adhesión Celular/inmunología , Endotelio Vascular/citología , Homocisteína/farmacología , Monocitos/citología , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Endotelio Vascular/metabolismo , Homocisteína/química , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Isomerismo , Leucemia Monocítica Aguda , Conformación Molecular , Monocitos/metabolismo , FN-kappa B/metabolismo , Nitrilos/farmacología , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Sulfonas/farmacología , Superóxidos/metabolismo , Venas Umbilicales/citología , Vasculitis/inmunología , Vasculitis/metabolismo
18.
Cell Mol Immunol ; 14(12): 986-996, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27086952

RESUMEN

Hepatitis C virus (HCV) infection is a major problem worldwide. HCV is not limited to liver disease but is frequently complicated by immune-mediated extrahepatic manifestations such as glomerulonephritis or vasculitis. A fatal complication of HCV-associated vascular disease is thrombosis. Polyriboinosinic:polyribocytidylic acid (poly (I:C)), a synthetic analog of viral RNA, induces a Toll-like receptor 3 (TLR3)-dependent arteriolar thrombosis without significant thrombus formation in venules in vivo. These procoagulant effects are caused by increased endothelial synthesis of tissue factor and PAI-1 without platelet activation. In addition to human umbilical endothelial cells (HUVEC), human mesangial cells (HMC) produce procoagulatory factors, cytokines and adhesion molecules after stimulation with poly (I:C) or HCV-containing cryoprecipitates from a patient with a HCV infection as well. Activated protein C (APC) is able to prevent the induction of procoagulatory factors in HUVEC and HMC in vitro and blocks the effects of poly (I:C) and HCV-RNA on the expression of cytokines and adhesion molecules in HMC but not in HUVEC. In vivo, protein C inhibits poly (I:C)-induced arteriolar thrombosis. Thus, endothelial cells are de facto able to actively participate in immune-mediated vascular thrombosis caused by viral infections. Finally, we provide evidence for the ability of protein C to inhibit TLR3-mediated arteriolar thrombosis caused by HCV infection.


Asunto(s)
Arterias/patología , Células Endoteliales/inmunología , Hepacivirus/inmunología , Hepatitis C/tratamiento farmacológico , Células Mesangiales/inmunología , Proteína C/uso terapéutico , Enfermedades Vasculares/tratamiento farmacológico , Vénulas/patología , Animales , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Poli I-C/administración & dosificación , Trombosis , Receptor Toll-Like 3/genética , Receptor Toll-Like 3/metabolismo
19.
Theranostics ; 7(2): 295-307, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28042335

RESUMEN

In the field of vascular gene therapy, targeting systems are promising advancements to improve site-specificity of gene delivery. Here, we studied whether incorporation of magnetic nanoparticles (MNP) with different magnetic properties into ultrasound sensitive microbubbles may represent an efficient way to enable gene targeting in the vascular system after systemic application. Thus, we associated novel silicon oxide-coated magnetic nanoparticle containing microbubbles (SO-Mag MMB) with lentiviral particles carrying therapeutic genes and determined their physico-chemical as well as biological properties compared to MMB coated with polyethylenimine-coated magnetic nanoparticles (PEI-Mag MMB). While there were no differences between both MMB types concerning size and lentivirus binding, SO-Mag MMB exhibited superior characteristics regarding magnetic moment, magnetizability as well as transduction efficiency under static and flow conditions in vitro. Focal disruption of lentiviral SO-Mag MMB by ultrasound within isolated vessels exposed to an external magnetic field decisively improved localized VEGF expression in aortic endothelium ex vivo and enhanced the angiogenic response. Using the same system in vivo, we achieved a highly effective, site-specific lentiviral transgene expression in microvessels of the mouse dorsal skin after arterial injection. Thus, we established a novel lentiviral MMB technique, which has great potential towards site-directed vascular gene therapy.


Asunto(s)
Vasos Sanguíneos/efectos de los fármacos , Sistemas de Liberación de Medicamentos , Terapia Genética/métodos , Vectores Genéticos , Lentivirus/genética , Nanopartículas de Magnetita/administración & dosificación , Microburbujas , Animales , Marcación de Gen/métodos , Ratones
20.
J Am Coll Cardiol ; 45(10): 1700-6, 2005 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-15893190

RESUMEN

OBJECTIVES: We investigated the role of SH2-domain containing phosphatase-1 (SHP-1) in endothelial reduced nicotinamide adenine dinucleotide (phosphate) (NAD[P]H)-oxidase-dependent oxidant production. BACKGROUND: Superoxide (O2*-) generation by endothelial NAD(P)H-oxidase promotes endothelial dysfunction and atherosclerosis. Signaling pathways that regulate NAD(P)H-oxidase activity are, however, poorly understood. METHODS: SH2-domain containing phosphatase-1 was inhibited using site-directed magnetofection of antisense oligodesoxynucleotides (AS-ODN) or short interfering ribonucleic acid (siRNA) in vitro in human umbilical vein endothelial cells (HUVEC) and in isolated hamster arteries; O2*- was measured by cytochrome c reduction in vitro. Activities of NAD(P)H-oxidase activity, phosphatidyl-inositol-3-kinase (PI3K), and SHP-1 were assessed by specific assays; Rac1 activation was assessed by a pull-down assay. RESULTS: Basal endothelial O2*- release was enhanced after inhibition of endothelial SHP-1 (p < 0.01), which could be prevented by specific inhibition of NAD(P)H-oxidase (p < 0.01); SHP-1 activity was high under basal conditions, further increased by vascular endothelial growth factor (10 ng/ml, p < 0.05), and abolished by SHP-1 AS-ODN treatment (p < 0.01), which also increased NAD(P)H-oxidase activity 3.3-fold (p < 0.01). Vascular endothelial growth factor also induced O2*- release (p < 0.01), which was even more enhanced when SHP-1 was knocked down (p < 0.05). The effect of SHP-1 was mediated by inhibition of PI3K/Rac1-dependent NAD(P)H-oxidase activation (p < 0.01); SHP-1 AS-ODN augmented tyrosine phosphorylation of the p85 regulatory subunit of PI3K (p < 0.05) and Rac1 activation. The latter was prevented by wortmannin, a blocker of PI3K. CONCLUSIONS: In HUVEC, SHP-1 counteracts basal and stimulated NAD(P)H-oxidase activity by negative regulation of PI3K-dependent Rac1 activation; SHP-1 thus seems to be an important part of endothelial antioxidative defense controlling the activity of the O2(*-)-producing NAD(P)H-oxidase.


Asunto(s)
Endotelio Vascular/fisiología , NADPH Oxidasas/antagonistas & inhibidores , Proteínas Tirosina Fosfatasas/fisiología , Superóxidos/metabolismo , Animales , Línea Celular , Cricetinae , Células Endoteliales/fisiología , Activación Enzimática/fisiología , Humanos , Técnicas In Vitro , Péptidos y Proteínas de Señalización Intracelular , NADPH Oxidasas/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteína Fosfatasa 1 , Proteína Tirosina Fosfatasa no Receptora Tipo 6 , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA