Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Part Fibre Toxicol ; 9: 32, 2012 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-22882971

RESUMEN

BACKGROUND: Respirable crystalline silica (silicon dioxide; SiO2, quartz) particles are known to induce chronic inflammation and lung disease upon long-term inhalation, whereas non-crystalline (amorphous) SiO2 particles in the submicrometre range are regarded as less harmful. Several reports have demonstrated that crystalline, but also non-crystalline silica particles induce IL-1ß release from macrophages via the NALP3-inflammasome complex (caspase-1, ASC and NALP3) in the presence of lipopolysaccharide (LPS) from bacteria. Our aim was to study the potential of different non-crystalline SiO2 particles from the nano- to submicro-sized range to activate IL-1ß responses in LPS-primed RAW264.7 macrophages and primary rat lung macrophages. The role of the NALP3-inflammasome and up-stream mechanisms was further explored in RAW264.7 cells. RESULTS: In the present study, we have shown that 6 h exposure to non-crystalline SiO2 particles in nano- (SiNPs, 5-20 nm, 50 nm) and submicro-sizes induced strong IL-1ß responses in LPS-primed mouse macrophages (RAW264.7) and primary rat lung macrophages. The primary lung macrophages were more sensitive to Si-exposure than the RAW-macrophages, and responded more strongly. In the lung macrophages, crystalline silica (MinUsil 5) induced IL-1ß release more potently than the non-crystalline Si50 and Si500, when adjusted to surface area. This difference was much less pronounced versus fumed SiNPs. The caspase-1 inhibitor zYVAD and RNA silencing of the NALP3 receptor reduced the particle-induced IL-1ß release in the RAW264.7 macrophages. Furthermore, inhibitors of phagocytosis, endosomal acidification, and cathepsin B activity reduced the IL-1ß responses to the different particles to a similar extent. CONCLUSIONS: In conclusion, non-crystalline silica particles in the nano- and submicro-size ranges seemed to induce IL-1ß release from LPS-primed RAW264.7 macrophages via similar mechanisms as crystalline silica, involving particle uptake, phagosomal leakage and activation of the NALP3 inflammasome. Notably, rat primary lung macrophages were more sensitive with respect to silica-induced IL-1ß release. The differential response patterns obtained suggest that silica-induced IL-1ß responses not only depend on the particle surface area, but on factors and/or mechanisms such as particle reactivity or particle uptake. These findings may suggest that bacterial infection via LPS may augment acute inflammatory effects of non-crystalline as well as crystalline silica particles.


Asunto(s)
Interleucina-1beta/metabolismo , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Nanopartículas/efectos adversos , Dióxido de Silicio/farmacología , Animales , Transporte Biológico , Línea Celular Transformada , Células Cultivadas , Inflamasomas/metabolismo , Lipopolisacáridos , Macrófagos/inmunología , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/metabolismo , Masculino , Ratones , Conformación Molecular , Nanopartículas/química , Nanopartículas/ultraestructura , Tamaño de la Partícula , Fagosomas/efectos de los fármacos , Fagosomas/inmunología , Fagosomas/metabolismo , Ratas , Ratas Endogámicas WKY , Dióxido de Silicio/efectos adversos , Dióxido de Silicio/química , Dióxido de Silicio/metabolismo , Propiedades de Superficie
2.
Toxicol Lett ; 208(3): 197-213, 2012 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-22108609

RESUMEN

Nanoparticles (NPs) occurring in the environment rapidly agglomerate and form particles of larger diameters. The extent to which this abates the effects of NPs has not been clarified. The motivation of this study was to examine how the agglomeration/aggregation state of silver (20nm and 200nm) and titanium dioxide (21nm) nanoparticles may affect the kinetics of cellular binding/uptake and ability to induce cytotoxic responses in THP1, HepG2 and A549 cells. Cellular binding/uptake, metabolic activation and cell death were assessed by the SSC flow cytometry measurements, the MTT-test and the propidium iodide assay. The three types of particles were efficiently taken up by the cells, decreasing metabolic activation and increasing cell death in all the cell lines. The magnitude of the studied endpoints depended on the agglomeration/aggregation state of particles, their size, time-point and cell type. Among the three cell lines tested, A549 cells were the most sensitive to these particles in relation to cellular binding/uptake. HepG2 cells showed a tendency to be more sensitive in relation to metabolic activation. THP-1 cells were the most resistant to all three types of particles in relation to all endpoints tested. Our findings suggest that particle features such as size and agglomeration status as well as the type of cells may contribute to nanoparticles biological impact.


Asunto(s)
Nanopartículas del Metal/toxicidad , Plata/toxicidad , Titanio/toxicidad , Muerte Celular/efectos de los fármacos , Endocitosis/efectos de los fármacos , Citometría de Flujo , Células Hep G2 , Humanos , Cinética , Nanopartículas del Metal/química , Microscopía Electrónica de Rastreo , Microscopía Electrónica de Transmisión , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Tamaño de la Partícula , Plata/química , Plata/farmacocinética , Estadísticas no Paramétricas , Titanio/química , Titanio/farmacocinética
3.
Toxicology ; 291(1-3): 65-72, 2012 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-22085606

RESUMEN

Serious concerns have been expressed about potential risks of engineered nanoparticles. Regulatory health risk assessment of such particles has become mandatory for the safe use of nanomaterials in consumer products and medicines; including the potential effects on reproduction and fertility, are relevant for this risk evaluation. In this study, we examined effects of silver particles of nano- (20nm) and submicron- (200nm) size, and titanium dioxide nanoparticles (TiO(2)-NPs; 21nm), with emphasis on reproductive cellular- and genotoxicity. Ntera2 (NT2, human testicular embryonic carcinoma cell line), and primary testicular cells from C57BL6 mice of wild type (WT) and 8-oxoguanine DNA glycosylase knock-out (KO, mOgg1(-/-)) genotype were exposed to the particles. The latter mimics the repair status of human testicular cells vs oxidative damage and is thus a suitable model for human male reproductive toxicity studies. The results suggest that silver nano- and submicron-particles (AgNPs) are more cytotoxic and cytostatic compared to TiO(2)-NPs, causing apoptosis, necrosis and decreased proliferation in a concentration- and time-dependent manner. The 200nm AgNPs in particular appeared to cause a concentration-dependent increase in DNA-strand breaks in NT2 cells, whereas the latter response did not seem to occur with respect to oxidative purine base damage analysed with any of the particles tested.


Asunto(s)
Citotoxinas/toxicidad , Mutágenos/toxicidad , Nanopartículas/toxicidad , Plata/toxicidad , Testículo/citología , Testículo/efectos de los fármacos , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Colorantes , Ensayo Cometa , Medios de Cultivo , Citocinas/biosíntesis , Daño del ADN , ADN Glicosilasas/genética , Células Germinativas/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Tamaño de la Partícula , Reproducción/efectos de los fármacos , Sales de Tetrazolio , Tiazoles , Titanio/toxicidad
4.
Ann Med ; 43(2): 90-103, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21039303

RESUMEN

Accumulating evidence suggests that inflammatory pathways play an essential role in all stages of atherogenesis. Inflammatory processes are not only involved in plaque progression, but seem also to play a critical role in plaque rupture. Members of the tumor necrosis factor (TNF) superfamiliy are potent regulators of inflammation and cell survival and consist of 20 ligands that signal through 29 different receptors. Several lines of evidence suggest that TNF-related molecules are involved in the development of acute coronary syndromes (ACS). Most, convincing evidence exists for CD40 ligand-CD40 interaction, but several other members of the TNF superfamily seem also to be involved in this immune-mediated promotion of plaque instability, including LIGHT, receptor activator of nuclear factor κB ligand, and TNF-α. These plaque destabilization pathways involve the bidirectional interaction between platelets and endothelial cells/monocytes, activation of vascular smooth muscle cells, and co-stimulatory effects on T cells, promoting inflammation, thrombus formation, matrix degradation, and apoptosis. TNF-related pathways could contribute to the non-resolving inflammation that characterizes atherosclerosis, representing pathogenic loops that are operating during plaque rupture and the development of ACS. These TNF-related molecules could also represent attractive new targets for therapy in this disorder.


Asunto(s)
Síndrome Coronario Agudo/fisiopatología , Receptores del Factor de Necrosis Tumoral/metabolismo , Factores de Necrosis Tumoral/metabolismo , Síndrome Coronario Agudo/tratamiento farmacológico , Animales , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/fisiopatología , Sistemas de Liberación de Medicamentos , Humanos , Inflamación/tratamiento farmacológico , Inflamación/fisiopatología , Ligandos , Transducción de Señal
5.
Scand Cardiovasc J ; 44(6): 359-66, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20799913

RESUMEN

OBJECTIVE: Notch signaling is crucial for cell-to-cell interaction during cardiovascular development and may influence differentiation, proliferation, and apoptotic events. We investigated whether Notch signaling is activated during myocardial remodeling in heart failure (HF). DESIGN: Myocardial gene expression and localization of Notch receptors (Notch1-4) and ligands (Jagged1-2, and Delta-like (Dll)-1 and 4) were investigated in rats with HF after induction of myocardial infarction and in humans with HF. RESULTS: All Notch receptors and ligands investigated and Notch intracellular domain (NICD) were present in rat and human myocardial tissue and in cardiomyocytes with differences in their relative expression levels and altered expression levels in failing vs. non-failing myocardium. In isolated rat cardiomyocytes, Notch3 and Notch4 appeared to be the major Notch receptors, and Notch3 and Notch4 mRNA levels and NICD-3 and -4 in cardiomyocytes were upregulated in chronic HF (p < 0.05), indicating increased Notch3 and Notch4 signaling. CONCLUSION: The Notch signaling system is present in the cardiomyocytes and activated in HF, indicating a role of Notch signaling during myocardial remodeling in HF.


Asunto(s)
Insuficiencia Cardíaca/patología , Infarto del Miocardio/patología , Miocardio/patología , Miocitos Cardíacos/patología , Receptores Notch , Transducción de Señal , Animales , Western Blotting , Progresión de la Enfermedad , Insuficiencia Cardíaca/genética , Inmunohistoquímica , Ligandos , Masculino , Infarto del Miocardio/genética , ARN Mensajero/genética , Ratas , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sístole , Factores de Tiempo
6.
Eur J Endocrinol ; 162(2): 267-73, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19926782

RESUMEN

OBJECTIVES: Thiazolidinediones (TZDs) reduce insulin resistance, but also have pleiotropic properties including effects on inflammation. The balance between protective and proatherogenic effects may differ in various patient populations. We studied the effect of rosiglitazone on inflammatory markers in patients with metabolic syndrome (MetSyn). METHODS: In a cross-over randomized controlled trial, 23 subjects with MetSyn were assigned to treatment with rosiglitazone that was uptitrated from 4 mg/day for 6 weeks followed by 8 mg/day for 6 weeks or matching placebo for 12 weeks, and then to the opposite treatment for 12 weeks. Plasma levels of inflammatory and metabolic markers were measured during follow-up. RESULTS: Our main findings were i) compared to placebo, rosiglitazone significantly decreased the plasma levels of the naturally occurring interleukin (IL)1 inhibitor, IL1 receptor antagonist (IL1Ra; P=0.001), potentially reflecting inflammatory effects on the IL1 system; ii) parallel to this, rosiglitazone decreased plasma levels of IL10 (P=0.029) further suggesting inflammatory effects; iii) rosiglitazone decreased uric acid levels (P=0.001), and monocyte chemoattractant protein-1 (P=0.05) and C-reactive protein (P=0.06) tended to be lower after rosiglitazone than placebo, suggesting potential pro- and anti-inflammatory effects simultaneously and iv) in vitro, rosiglitazone enhanced IL1Ra and decreased IL1beta in THP-1 monocytes, illustrating the complex effects of these medications, potentially exhibiting anti-inflammatory effects on the IL1 system in certain tissues or cells at least at certain concentrations. CONCLUSION: Our findings suggest inflammatory effects on the IL1 system during rosiglitazone therapy in MetSyn. However, anti-inflammatory effects were also observed, and the net effect of TZDs in MetSyn should be further investigated.


Asunto(s)
Hipoglucemiantes/administración & dosificación , Proteína Antagonista del Receptor de Interleucina 1/metabolismo , Síndrome Metabólico/tratamiento farmacológico , Síndrome Metabólico/metabolismo , PPAR gamma/agonistas , Tiazolidinedionas/administración & dosificación , Adulto , Anciano , Biomarcadores , Estudios Cruzados , Regulación hacia Abajo/efectos de los fármacos , Femenino , Estudios de Seguimiento , Humanos , Hipoglucemiantes/efectos adversos , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Masculino , Síndrome Metabólico/inmunología , Persona de Mediana Edad , Rosiglitazona , Tiazolidinedionas/efectos adversos
7.
Thromb Haemost ; 102(4): 704-10, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19806256

RESUMEN

Activated platelets release a wide range of inflammatory mediators, including members of the tumour necrosis factor (TNF) superfamily (e.g. CD40 ligand [CD40L] and LIGHT). Such platelet-mediated inflammation could be involved in atherogenesis and plaque destabilisation. In the present study we investigated whether APRIL, another member of the TNF superfamily that has been detected in megakaryocytes, could be released from platelets upon activation. The release of APRIL was studied in thrombin receptor (SFLLRN) activated platelets, and the expression of APRIL was examined in plasma and within the atherosclerotic lesion in patients with carotid and coronary atherosclerosis. Upon SFLLRN activation, there was a gradual release of APRIL, reaching maximum after 90 minutes. While this pattern is similar to that of CD40L and LIGHT, the release of APRIL was quite differently regulated. Thus, prostaglandin E1, but not inhibitors of metal-dependent proteases and actin polymerisation or the lack of GP IIb/IIIa, blocks APRIL release in activated platelets. With relevance to atherogenesis, we found that patients with coronary artery disease (n=80) had raised plasma levels of APRIL as compared with controls (n=20), and APRIL immunoreactivity was detected in aggregated platelets within the ruptured plaque in patients with myocardial infarction and within macrophages in symptomatic carotid plaques. In conclusion, activated platelets release significant amounts of APRIL in a long-lasting manner, differently regulated than the gradual release of other platelet-derived TNF superfamily ligands. The enhanced expression of APRIL in atherosclerotic disorders, both systemically and within the lesion, may suggest a potential involvement of APRIL in atherogenesis.


Asunto(s)
Plaquetas/metabolismo , Enfermedad de la Arteria Coronaria/inmunología , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/biosíntesis , Anciano , Alprostadil/inmunología , Alprostadil/metabolismo , Apoptosis , Plaquetas/inmunología , Plaquetas/patología , Ligando de CD40/inmunología , Ligando de CD40/metabolismo , Arterias Carótidas/metabolismo , Arterias Carótidas/patología , Diferenciación Celular , Proliferación Celular , Enfermedad de la Arteria Coronaria/sangre , Enfermedad de la Arteria Coronaria/genética , Enfermedad de la Arteria Coronaria/patología , Enfermedad de la Arteria Coronaria/fisiopatología , Femenino , Regulación de la Expresión Génica , Humanos , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Persona de Mediana Edad , Activación Plaquetaria , Receptores de Trombina/inmunología , Receptores de Trombina/metabolismo , Transducción de Señal , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/inmunología , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/inmunología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo
8.
Arterioscler Thromb Vasc Biol ; 29(8): 1228-34, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19498175

RESUMEN

OBJECTIVE: Based on the emerging importance of the wingless (Wnt) pathways in inflammation and vascular biology, we hypothesized a role for Dickkopf-1 (DKK-1), a major modulator of Wnt signaling, in atherogenesis and plaque destabilization. METHODS AND RESULTS: We report increased levels of DKK-1 in experimental (ApoE(-/-) mice) and clinical (patients with coronary artery disease [n=80] and patients with carotid plaque [n=47]) atherosclerosis, both systemically (serum) and within the lesion, with particularly high levels in advanced and unstable disease. We identified platelets as an important cellular source of DKK-1 as shown by in vitro experiments and by immunostaining of thrombus material obtained at the site of plaque rupture in patients with acute ST-elevation myocardial infarction, with strong immunoreactivity in platelet aggregates. Our in vitro experiments identified a role for platelet- and endothelial-derived DKK-1 in platelet-dependent endothelial activation, promoting enhanced release of inflammatory cytokines. These inflammatory effects of DKK-1 involved inhibition of the Wnt/beta-catenin pathway and activation of nuclear factor kappaB. CONCLUSIONS: Our findings identify DKK-1 as a novel mediator in platelet-mediated endothelial cell activation. The demonstration of enhanced DKK-1 expression within advanced carotid plaques may suggest that this DKK-1-driven inflammatory loop could be operating within the atherosclerotic lesion.


Asunto(s)
Aterosclerosis/metabolismo , Plaquetas/metabolismo , Endotelio Vascular/metabolismo , Regulación de la Expresión Génica , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Animales , Aterosclerosis/patología , Plaquetas/patología , Western Blotting , Arterias Carótidas/metabolismo , Arterias Carótidas/patología , Células Cultivadas , Vasos Coronarios/metabolismo , Vasos Coronarios/patología , Modelos Animales de Enfermedad , Endotelio Vascular/patología , Humanos , Ratones , Ratones Endogámicos C57BL , Pronóstico
9.
Circ Res ; 104(1): 60-8, 2009 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-19023130

RESUMEN

The interaction between inflammatory cytokines and endothelial cells is a critical step in atherogenesis leading to endothelial dysfunction and inflammation. We have previously reported that the tumor necrosis factor superfamily member LIGHT could be involved in atherogenesis through its ability to promote vascular inflammation. In the present study we identified proteinase-activated receptor (PAR)-2 as an inflammatory mediator that was markedly enhanced by LIGHT in endothelial cells. We also found that LIGHT acted synergistically with PAR-2 activation to promote enhanced release of the proatherogenic chemokines interleukin-8 and monocyte chemoattractant protein-1, underscoring that the interaction between LIGHT and PAR-2 is biologically active, promoting potent inflammatory effects. We showed that the LIGHT-mediated upregulation of PAR-2 in endothelial cells is mediated through the HVEM receptor, involving Jun N-terminal kinase signaling pathways. A LIGHT-mediated upregulation of PAR-2 mRNA levels was also found in human monocytes when these cells were preactivated by tumor necrosis factor alpha. We have previously demonstrated increased plasma levels of LIGHT in unstable angina patients, and here we show a similar pattern for PAR-2 expression in peripheral blood monocytes. We also found that LIGHT, LIGHT receptors, and PAR-2 showed enhanced expression, and, to some degree, colocalization in endothelial cells and macrophages, in the atherosclerotic plaques of ApoE(-/-) mice, suggesting that the inflammatory interaction between LIGHT and PAR-2 also may be operating in vivo within an atherosclerotic lesion. Our findings suggest that LIGHT/PAR-2-driven inflammation could be a pathogenic loop in atherogenesis potentially representing a target for therapy in this disorder.


Asunto(s)
Aterosclerosis/etiología , Células Endoteliales/metabolismo , Endotelio Vascular/patología , Receptor PAR-2/fisiología , Miembro 14 de Receptores del Factor de Necrosis Tumoral/fisiología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología , Vasculitis/metabolismo , Anciano , Angina de Pecho/metabolismo , Angina de Pecho/patología , Angina Inestable/metabolismo , Angina Inestable/patología , Animales , Aterosclerosis/metabolismo , Aterosclerosis/patología , Células Cultivadas/metabolismo , Quimiocina CCL2/metabolismo , Células Endoteliales/patología , Femenino , Regulación de la Expresión Génica , Humanos , Interleucina-8/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Óxido Nítrico Sintasa de Tipo III/metabolismo , Receptor PAR-2/agonistas , Proteínas Recombinantes de Fusión/fisiología , Transducción de Señal/fisiología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética , Vasculitis/complicaciones , Vasculitis/patología
10.
Curr Atheroscler Rep ; 10(3): 236-43, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18489852

RESUMEN

Although the pathogenic role of T cells in atherogenesis is well established, the function of the various T-cell subsets is far from clear. Whereas activation of the T-helper type 1 (Th1) subset promotes inflammatory and proatherogenic responses and activation of Th2 cells mediates both proatherogenic and antiatherogenic effects, the newly discovered regulatory T-cell subset seems to attenuate atherogenesis. However, the dynamics of T-cell response within the plaque are still poorly understood, and both antigen-dependent and antigen-independent stimuli may be involved in the expansion of T cells in atherosclerotic plaques. Nevertheless, the different nature of the various T-cell subsets and their complex role in atherogenesis underscore the need for future research in this field of atheroimmunology. This research is not only of interest for the basic research field, but may also have relevance for clinical cardiology, potentially leading to new targets for therapy in atherosclerotic disorders.


Asunto(s)
Aterosclerosis/inmunología , Citocinas/inmunología , Linfocitos T/inmunología , Citocinas/metabolismo , Humanos , Interleucina-17/metabolismo , Activación de Linfocitos/fisiología , Macrófagos/metabolismo , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
11.
Eur J Heart Fail ; 10(4): 352-9, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18353719

RESUMEN

BACKGROUND: Clinical and experimental studies suggest a pathogenic role for inflammation in chronic heart failure (HF). LIGHT is a member of the tumour necrosis factor superfamily involved in innate and adaptive immune responses. AIMS: We sought to investigate a potential pathogenic role of LIGHT in chronic HF. METHODS: We used various clinical and experimental approaches including studies in post-infarction HF rats and in vitro studies of endothelial cells and peripheral blood mononuclear cells (PBMC). RESULTS: Our main findings were: (i) LIGHT and its receptors (i.e., HVEM and lymphotoxin-beta receptor) were regulated during experimental HF, with strong expression in the infarcted area accompanied by up-regulation of HVEM in cardiomyocytes and endothelial cells also in the non-ischaemic part of the left ventricle. (ii) Patients with chronic HF had significantly increased expression of LIGHT on CD3(+) T-cells accompanied by increased expression of HVEM on monocytes and within the failing myocardium. (iii) LIGHT induced interleukin (IL)-6 expression in endothelial cells. In HF patients, but not in healthy controls, such an IL-6-inducing effect was also seen in LIGHT activated PBMC. CONCLUSION: Our findings in both clinical and experimental HF may suggest a role for LIGHT signalling pathways in the progression of chronic HF involving IL-6-related mechanisms.


Asunto(s)
Modelos Animales de Enfermedad , Insuficiencia Cardíaca/patología , Receptor beta de Linfotoxina/metabolismo , Miembro 14 de Receptores del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal/fisiología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo , Adulto , Anciano , Animales , Complejo CD3/metabolismo , Endotelio Vascular/patología , Femenino , Ventrículos Cardíacos/patología , Humanos , Técnicas In Vitro , Interleucina-6/metabolismo , Masculino , Persona de Mediana Edad , Monocitos/metabolismo , Infarto del Miocardio/patología , Miocitos Cardíacos/patología , Ratas , Ratas Wistar , Linfocitos T/patología , Regulación hacia Arriba/fisiología
12.
Cardiovasc Res ; 79(1): 195-203, 2008 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-18339644

RESUMEN

AIMS: CXC ligand 16 (CXCL16) may be involved in inflammation and lipid metabolism, and we hypothesized a role for this chemokine in coronary artery disease (CAD). METHODS AND RESULTS: We performed clinical studies in CAD patients as well as experimental studies in cells with relevance to atherogenesis [i.e. endothelial cells, vascular smooth muscle cells (SMC), and peripheral blood mononuclear cells (PBMC)]. We also examined the ability of HMG-CoA reductase inhibitors (statins) to modulate CXCL16 levels both in vivo and in vitro. Our main findings were: (i) patients with stable (n = 40) and unstable (n = 40) angina had elevated plasma levels of CXCL16 compared with controls (n = 20); (ii) low-dose simvastatin (20 mg qd, n = 15) and high-dose atorvastatin (80 mg qd, n = 9) down-regulated plasma levels of CXCL16 during 6 months of therapy; (iii) in vitro, atorvastatin significantly decreased the interleukin (IL)-1beta-mediated release of CXCL16 from PBMC and endothelial cells; (iv) attenuating effect of atorvastatin on the IL-1beta-mediated release of CXCL16 in PBMC seems to involve post-transcriptional modulation as well as down-regulation of CXCL16 release through inhibition of the protease a disintegrin and metalloproteinase 10 (ADAM10); (v) soluble CXCL16 increased the release of IL-8, monocyte chemoattractant peptide 1, and matrix metalloproteinases in vascular SMC and increased the release of IL-8 and monocyte chemoattractant peptide 1 in PBMC, with particularly enhancing effects in cells from CAD patients. CONCLUSION: Our findings suggest that soluble CXCL16 could be linked to atherogenesis not only as a marker of inflammation, but also as a potential inflammatory mediator.


Asunto(s)
Quimiocinas CXC/metabolismo , Enfermedad de la Arteria Coronaria/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Receptores Depuradores/metabolismo , Anciano , Atorvastatina , Estudios de Casos y Controles , Células Cultivadas , Quimiocina CCL2/metabolismo , Quimiocina CXCL16 , Quimiocinas CXC/genética , Enfermedad de la Arteria Coronaria/tratamiento farmacológico , Enfermedad de la Arteria Coronaria/patología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Femenino , Ácidos Heptanoicos/farmacología , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Interleucina-8/metabolismo , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/patología , Masculino , Persona de Mediana Edad , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Pirroles/farmacología , Receptores Depuradores/genética , Simvastatina/farmacología
13.
Arterioscler Thromb Vasc Biol ; 28(5): 1005-11, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18276907

RESUMEN

OBJECTIVE: We examined the role of the CXCR2 ligand growth-related oncogene (GRO) alpha in human atherosclerosis. METHODS AND RESULTS: GROalpha levels were examined by enzyme immunoassay, real-time quantitative RT-PCR, and cDNA microarrays. The in vitro effect of statins on GROalpha was examined in endothelial cells and THP-1 macrophages. Our main findings were: (1) GROalpha was among the 10 most differentially expressed transcripts comparing peripheral blood mononuclear cells (PBMCs) from patients with coronary artery disease (CAD) and healthy controls. (2) Both patients with stable (n=41) and particularly those with unstable (n=47) angina had increased plasma levels of GROalpha comparing controls (n=20). (3) We found increased expression of GROalpha within symptomatic carotid plaques, located to macrophages and endothelial cells. (4) GROalpha enhanced the release of matrix metalloproteinases in vascular smooth muscle cells, and increased the binding of acetylated LDL in macrophages. (5) Atorvastatin downregulated GROalpha levels as shown both in vitro in endothelial cells and macrophages and in vivo in PBMCs from CAD patients. (6) The effect on GROalpha in endothelial cells involved increased storage and reduced secretion of GROalpha. CONCLUSIONS: GROalpha could be involved in atherogenesis and plaque destabilization, potentially contributing to inflammation, matrix degradation, and lipid accumulation within the atherosclerotic lesion.


Asunto(s)
Estenosis Carotídea/metabolismo , Quimiocina CXCL1/metabolismo , Enfermedad de la Arteria Coronaria/metabolismo , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Angina Inestable/metabolismo , Angina Inestable/patología , Aorta/metabolismo , Aorta/patología , Estenosis Carotídea/patología , Estudios de Casos y Controles , Células Cultivadas , Quimiocina CXCL1/genética , Enfermedad de la Arteria Coronaria/patología , Regulación hacia Abajo , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Inhibidores Enzimáticos/farmacología , Femenino , Humanos , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/patología , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Metaloproteinasa 1 de la Matriz/metabolismo , Metaloproteinasa 3 de la Matriz/metabolismo , Persona de Mediana Edad , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Venas Umbilicales/metabolismo , Venas Umbilicales/patología
14.
Am J Respir Crit Care Med ; 177(2): 202-7, 2008 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-17962639

RESUMEN

RATIONALE: Thrombus formation and inflammation are involved in the pathogenesis of pulmonary arterial hypertension (PAH), and LIGHT (Lymphotoxin-like Inducible protein that competes with Glycoprotein D for Herpesvirus entry mediator on T lymphocytes) has been shown to promote vascular inflammation. OBJECTIVES: We sought to investigate the role of the tumor necrosis factor superfamily ligand LIGHT in the pathogenesis of PAH. METHODS: We studied 73 patients with severe PAH and 10 control subjects. LIGHT and pro- and antithrombotic markers were assessed by enzyme immunoassays. MEASUREMENTS AND MAIN RESULTS: (1) Patients with idiopathic PAH (n = 21), patients with PAH related to risk factors or associated conditions (n = 31), and those with chronic thromboembolic PAH (n = 21) all had raised serum levels of LIGHT compared with control subjects (n = 10). (2) LIGHT levels in femoral artery were significantly related to mortality in the patients with PAH. (3) Immunostaining of LIGHT and its receptors was seen in alveolar macrophages, vascular smooth muscle cells, and endothelial cells in lungs from patients with PAH. (4) Thirteen patients received prostacyclin infusion (3 mo), and all showed hemodynamic improvement, accompanied by decreased LIGHT levels. (5) Prostacyclin abolished the release of LIGHT from activated platelets in vitro, suggesting that the decrease in LIGHT during prostacyclin therapy could involve direct effects on platelets. (6) LIGHT increased tissue factor and plasminogen activator inhibitor type 1 and decreased thrombomodulin levels in endothelial cells, inducing a prothrombotic state in these cells. CONCLUSIONS: Our findings suggest prothrombotic effects of LIGHT in PAH involving endothelium-related mechanisms, potentially contributing to the progression of this disorder.


Asunto(s)
Hipertensión Pulmonar/etiología , Trombosis/etiología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología , Biomarcadores/sangre , Progresión de la Enfermedad , Humanos , Ligandos , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/análisis , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/sangre , Venas Umbilicales/citología
16.
Arterioscler Thromb Vasc Biol ; 27(3): 614-20, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17170367

RESUMEN

OBJECTIVE: Based on their role in T-cell homing into nonlymphoid tissue, we examined the role of the homeostatic chemokines CCL19 and CCL21 and their common receptor CCR7 in coronary artery disease (CAD). METHODS AND RESULTS: We performed studies in patients with stable (n=40) and unstable (n=40) angina and healthy controls (n=20), in vitro studies in T-cells and macrophages, and studies in apolipoprotein-E-deficient (ApoE-/-) mice and human atherosclerotic carotid plaques. We found increased levels of CCL19 and CCL21 within the atherosclerotic lesions of the ApoE-/- mice, in human atherosclerotic carotid plaques, and in plasma of CAD patients. Whereas strong CCR7 expression was seen in T-cells from murine and human atherosclerotic plaques, circulating T-cells from angina patients showed decreased CCR7 expression. CCL19 and CCL21 promoted an inflammatory phenotype in T-cells and macrophages and increased matrix metalloproteinase (MMP) and tissue factor levels in the latter cell type. Although aggressive statin therapy increased CCR7 and decreased CCL19/CCL21 levels in peripheral blood from CAD patients, conventional therapy did not. CONCLUSIONS: The abnormal regulation of CCL19 and CCL21 and their common receptor in atherosclerosis could contribute to disease progression by recruiting T-cells and macrophages to the atherosclerotic lesions and by promoting inflammatory responses in these cells.


Asunto(s)
Quimiocinas CC/metabolismo , Enfermedad Coronaria/sangre , Enfermedad Coronaria/tratamiento farmacológico , Ácidos Heptanoicos/uso terapéutico , Pirroles/uso terapéutico , Simvastatina/uso terapéutico , Angioplastia Coronaria con Balón/métodos , Animales , Apolipoproteínas E/deficiencia , Aterosclerosis/metabolismo , Aterosclerosis/patología , Aterosclerosis/prevención & control , Atorvastatina , Biopsia con Aguja , Células Cultivadas , Quimiocina CCL19 , Quimiocina CCL21 , Quimiocinas CC/genética , Enfermedad Coronaria/patología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Regulación de la Expresión Génica , Humanos , Inmunohistoquímica , Técnicas In Vitro , Leucocitos Mononucleares , Ratones , Ratones Transgénicos , Pronóstico , ARN Mensajero/análisis , Receptores CCR7 , Receptores de Quimiocina/metabolismo , Valores de Referencia , Factores de Riesgo , Sensibilidad y Especificidad , Resultado del Tratamiento
17.
J Am Coll Cardiol ; 48(8): 1591-9, 2006 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-17045893

RESUMEN

OBJECTIVES: We sought to investigate the role of the CXC chemokine neutrophil-activating peptide-2 (NAP-2) in atherogenesis and plaque destabilization. BACKGROUND: Chemokines are involved in atherogenesis, but the role of NAP-2 in atherosclerotic disorders is unclear. Based on its potential pro-atherogenic properties, we hypothesized a pathogenic role for NAP-2 in coronary artery disease. METHODS: We tested this hypothesis by differential experimental approaches including studies in patients with stable (n = 40) and unstable angina (n = 40) and healthy control subjects (n = 20). RESULTS: The following results were discovered: 1) patients with stable, and particularly those with unstable, angina had markedly raised plasma levels of NAP-2 compared with control subjects, accompanied by increased expression of CXC receptor 2 in monocytes; 2) platelets, but also peripheral blood mononuclear cells (PBMCs), released large amounts of NAP-2 upon stimulation, with a particularly prominent PBMC response in unstable angina; 3) NAP-2 protein was detected in macrophages and smooth muscle cells of atherosclerotic plaques and in monocytes and platelets of coronary thrombi; 4) in vitro, recombinant and platelet-derived NAP-2 increased the expression of adhesion molecules and chemokines in endothelial cells; and 5) whereas aspirin reduced plasma levels of NAP-2, statin therapy increased NAP-2 with stimulating effects both on platelets and leukocytes. CONCLUSIONS: Our findings suggest that NAP-2 has the potential to induce inflammatory responses within the atherosclerotic plaque. By its ability to promote leukocyte and endothelial cell activation, such a NAP-2-driven inflammation could promote plaque rupture and acute coronary syndromes.


Asunto(s)
Enfermedad Coronaria/sangre , beta-Tromboglobulina/metabolismo , Enfermedad Aguda , Angina de Pecho/sangre , Angina Inestable/sangre , Arterias , Aspirina/farmacología , Plaquetas/metabolismo , Moléculas de Adhesión Celular/metabolismo , Células Cultivadas , Quimiocinas/metabolismo , Quimiotaxis/efectos de los fármacos , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Arteriosclerosis Intracraneal/metabolismo , Arteriosclerosis Intracraneal/patología , Macrófagos/metabolismo , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/metabolismo , Recuento de Plaquetas , Receptores de Péptidos/sangre , Receptores de Péptidos/metabolismo , Proteínas Recombinantes/farmacología , Síndrome , Linfocitos T/metabolismo , Trombosis/sangre , Vasculitis/etiología , beta-Tromboglobulina/farmacología
18.
Arterioscler Thromb Vasc Biol ; 26(4): 857-63, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16424351

RESUMEN

OBJECTIVE: Based on its role in inflammation and matrix degradation, we hypothesized a role for osteoprotegerin (OPG), RANK, and RANK ligand (RANKL) in coronary artery disease. METHODS AND RESULTS: We examined the expression of various members of the OPG/RANKL/RANK axis in patients with stable and unstable angina and in the atherosclerotic lesions of apolipoprotein E-deficient (apoE(-/-)) mice. Our findings were: (1) Serum levels of OPG were raised in patients with unstable angina (n=40), but not in those with stable angina (n=40), comparing controls (n=20); (2) mRNA levels of RANKL were increased in T-cells in unstable angina patients accompanied by increased expression of RANK in monocytes; (3) strong immunostaining of OPG/RANKL/RANK was seen within thrombus material obtained at the site of plaque rupture during acute myocardial infarction; (4) OPG/RANKL/RANK was expressed in the atherosclerotic plaques of apoE(-/-) mice, with RANKL located specifically to the plaques; and (5) RANKL enhanced the release of monocyte chemoattractant peptide-1 in mononuclear cells from unstable angina patients, and promoted matrix metalloproteinase (MMP) activity in vascular smooth muscle cells. CONCLUSIONS: We show enhanced expression of the OPG/RANKL/RANK system both in clinical and experimental atherosclerosis, with enhanced T-cell expression of RANKL as an important feature of unstable disease.


Asunto(s)
Angina Inestable/metabolismo , Aterosclerosis/metabolismo , Proteínas Portadoras/metabolismo , Glicoproteínas/sangre , Glicoproteínas/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores Citoplasmáticos y Nucleares/sangre , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores del Factor de Necrosis Tumoral/sangre , Receptores del Factor de Necrosis Tumoral/metabolismo , Linfocitos T/metabolismo , Adulto , Anciano , Angina Inestable/inmunología , Angina Inestable/patología , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/inmunología , Aterosclerosis/patología , Línea Celular , Femenino , Humanos , Masculino , Metaloproteinasas de la Matriz/metabolismo , Ratones , Ratones Noqueados , Persona de Mediana Edad , Monocitos/metabolismo , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Osteoprotegerina , Ligando RANK , Receptor Activador del Factor Nuclear kappa-B , Rotura Espontánea/inmunología , Rotura Espontánea/metabolismo , Linfocitos T/patología
19.
Arterioscler Thromb Vasc Biol ; 26(1): 200-5, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16254204

RESUMEN

OBJECTIVE: Increasing data support the involvement of chemokines in atherogenesis. However, although several studies have shown increased chemokine levels in adult patients, the literature is virtually devoid of data on chemokines in children with hypercholesterolemia. METHODS AND RESULTS: We examined the gene expression of chemokines in peripheral blood mononuclear cells (PBMCs) from clinically healthy children with and without heterozygous familial hypercholesterolemia (FH). Our main findings were: (1) compared with healthy controls, PBMCs from FH children showed significantly higher mRNA levels of RANTES, but not of the other examined chemokines; (2) an opposite pattern was seen in adult FH subjects, with markedly enhanced expression of macrophage inflammatory peptide-1alpha, but not of RANTES; (3) this increased gene expression of RANTES in PBMCs from FH children seemed to reflect enhanced RANTES expression in monocytes but not in T cells; (4) FH children also had raised serum levels of neopterin, additionally suggesting monocyte/macrophage activation in these children; and (5) PBMCs from both FH children and controls showed enhanced release of interleukin 8 on RANTES stimulation in vitro. CONCLUSIONS: Our findings support a role of inflammation also in the early stages of atherogenesis possibly involving monocyte-derived RANTES as an important mediator.


Asunto(s)
Quimiocina CCL5/genética , Quimiocina CCL5/metabolismo , Hiperlipoproteinemia Tipo II/genética , Hiperlipoproteinemia Tipo II/inmunología , Adolescente , Adulto , Aterosclerosis/genética , Aterosclerosis/inmunología , Aterosclerosis/metabolismo , Células Cultivadas , Chaperonina 60/farmacología , Quimiocina CCL4 , Niño , Femenino , Heterocigoto , Humanos , Hiperlipoproteinemia Tipo II/metabolismo , Interleucina-8/genética , Interleucina-8/metabolismo , Lipoproteínas LDL/farmacología , Proteínas Inflamatorias de Macrófagos/genética , Proteínas Inflamatorias de Macrófagos/metabolismo , Masculino , Persona de Mediana Edad , Monocitos/citología , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Neopterin/sangre , ARN Mensajero/análisis , Linfocitos T/citología , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Regulación hacia Arriba/inmunología
20.
Arterioscler Thromb Vasc Biol ; 25(12): 2567-72, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16224053

RESUMEN

OBJECTIVE: Recent data derived primarily from studies in animal models suggest that fractalkine (CX3CL1) and its cognate receptor, CX3CR1, play a role in atherogenesis. We, therefore, hypothesized that enhanced CX3CL1/CX3CR1 expression may promote atherogenesis in patients with coronary artery disease (CAD). METHODS AND RESULTS: We examined the plasma levels of CX3CL1 and CX3CR1 expression in peripheral blood mononuclear cells (PBMC) in various CAD populations (30 patients with previous myocardial infarction, 40 patients with stable angina, 40 patients with unstable angina, and a total of 35 controls) and used various experimental approaches to characterize CX3CL1-mediated leukocyte responses. We found that the plasma levels of CX3CL1 are greatly increased in CAD, particularly in unstable disease. The parallel increase of CX3CR1 expression in PBMC was predominantly attributable to an expansion of the (CX3CR1+)(CD3+)(CD8+) T cell subset and was associated with enhanced chemotactic, adhesive, and inflammatory responses to CX3CL1. Statin therapy for 6 months reduced the expression of CX3CL1 and CX3CR1, reaching statistical significance for both parameters only during aggressive (atorvastatin, 80 mg qd) but not conventional (simvastatin, 20 mg qd) therapy. Consequently, the functional responses of the PBMC to CX3CL1 including migration, adhesion, and secretion of interleukin-8 were attenuated by the treatments. CONCLUSIONS: Our results suggest that the CX3CL1/CX3CR1 dyad may contribute to atherogenesis and plaque destabilization in human CAD.


Asunto(s)
Quimiocinas CX3C/sangre , Enfermedad de la Arteria Coronaria/tratamiento farmacológico , Enfermedad de la Arteria Coronaria/fisiopatología , Ácidos Heptanoicos/administración & dosificación , Inhibidores de Hidroximetilglutaril-CoA Reductasas/administración & dosificación , Proteínas de la Membrana/sangre , Proteínas de la Membrana/genética , Pirroles/administración & dosificación , Receptores de Quimiocina/genética , Angina Inestable/tratamiento farmacológico , Angina Inestable/metabolismo , Angina Inestable/fisiopatología , Atorvastatina , Receptor 1 de Quimiocinas CX3C , Adhesión Celular/efectos de los fármacos , Células Cultivadas , Quimiocina CX3CL1 , Quimiocinas CX3C/genética , Quimiocinas CX3C/farmacología , Quimiotaxis/efectos de los fármacos , LDL-Colesterol/sangre , Enfermedad de la Arteria Coronaria/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/fisiología , Expresión Génica/efectos de los fármacos , Expresión Génica/fisiología , Humanos , Interleucina-8/metabolismo , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Proteínas de la Membrana/farmacología , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/metabolismo , Infarto del Miocardio/fisiopatología , Simvastatina/administración & dosificación , Venas Umbilicales/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...