Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
2.
J Immunol ; 177(11): 8266-72, 2006 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-17114505

RESUMEN

IL-22 is a recently discovered cytokine of the IL-10 family that binds to a class II cytokine receptor composed of IL-22R1 and IL-10R2(c) and influences a variety of immune reactions. As IL-22 has also been shown to modulate cell cycle and proliferation mediators such as ERK1/2 and JNK, we studied the role of IL-22 in proliferation, apoptosis, and cell cycle regulation in EMT6 murine breast cancer cells in vitro and in vivo. In this study, we report that murine breast cancer cells express functional IL-22R as indicated by RT-PCR studies, immunoblotting, and STAT3 activation assays. Importantly, IL-22 exposure of EMT6 cells resulted in decreased levels of phosphorylated ERK1/2 and AKT protein kinases, indicating an inhibitory effect of IL-22 on signaling pathways promoting cell proliferation. Furthermore, IL-22 induced a cell cycle arrest of EMT6 cells in the G(2)-M phase. IL-22 reduced EMT6 cell numbers and the proliferation rate by approximately 50% as measured by [(3)H]thymidine incorporation. IL-22 treatment of EMT6 tumor-bearing mice lead to a decreased tumor size and a reduced tumor cell proliferation in vivo, as determined by 3'-deoxy-3'-fluorothymidine-positron emission tomography scans. Interestingly, IL-22 did not induce apoptosis, as determined in annexin V binding assay and caspase-3 activation assay and had no effect on angiogenesis in vivo. In conclusion, our results indicate that IL-22 reduced tumor growth by inhibiting signaling pathways such as ERK1/2 and AKT phosphorylation that promote tumor cell proliferation in EMT6 cells. Therefore, IL-22 may play a role in the control of tumor growth and tumor progression.


Asunto(s)
Neoplasias de la Mama/metabolismo , Ciclo Celular/fisiología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Interleucinas/metabolismo , Proteína Oncogénica v-akt/metabolismo , Transducción de Señal/fisiología , Animales , Apoptosis/fisiología , Western Blotting , Línea Celular Tumoral , Proliferación Celular , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Expresión Génica , Ratones , Fosforilación , ARN Mensajero/análisis , Receptores de Interleucina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Interleucina-22
3.
Am J Physiol Heart Circ Physiol ; 290(6): H2178-86, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16473961

RESUMEN

Induction of apoptosis represents a potential reaction of endothelial cells (ECs) after injury of the vascular endothelium. Beneficial effects of n-3 polyunsaturated fatty acids (PUFAs) in vascular diseases are widely recognized although the responsible mechanisms are not fully understood. Because it is not known whether PUFAs modulate EC apoptosis, we investigated the effects of n-3 and n-6 PUFAs on 4-hydroxynonenal (HNE)-induced EC apoptosis by annexin V staining and caspase-3 activation assays. Pretreatment with the n-3 fatty acid docosahexaenoic acid (DHA) reduced HNE-induced EC apoptosis. DHA-treated cells did not show the pronounced drop in intracellular GSH after HNE exposure seen in vehicle- or n-6 arachidonic acid-treated cells. This is most likely due to increased GSH levels in DHA-treated cells. Furthermore, DHA pretreatment increased ciap1 mRNA levels and transfection of cIAP1 small interfering RNA abolished the protective effect of DHA in HNE-induced apoptosis in HUVECs. Thus pretreatment of HUVECs with DHA reduces HNE-induced oxidative stress and apoptosis, and the protective effects of DHA seem to be dependent on cIAP1. The results provide a possible new mechanism for the atheroprotective effects of n-3 fatty acids in vascular disease.


Asunto(s)
Apoptosis/efectos de los fármacos , Ácidos Docosahexaenoicos/farmacología , Células Endoteliales/efectos de los fármacos , Proteínas Inhibidoras de la Apoptosis/biosíntesis , Aldehídos/antagonistas & inhibidores , Aldehídos/toxicidad , Anexina A5/metabolismo , Western Blotting , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Citocromos c/metabolismo , Citometría de Flujo , Glutatión/metabolismo , Humanos , Proteínas Inhibidoras de la Apoptosis/antagonistas & inhibidores , Proteínas Inhibidoras de la Apoptosis/genética , Estrés Oxidativo/efectos de los fármacos , ARN Mensajero/biosíntesis , ARN Interferente Pequeño/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Estrés Fisiológico/patología , Superóxidos/metabolismo , Transfección
4.
FASEB J ; 19(10): 1293-5, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15939736

RESUMEN

Nuclear factor-kappaB (NF-kappaB)-mediated vascular inflammation is a prominent characteristic of atherogenesis and restenosis. We noted that angioplastic injury to carotid artery elicited two phases of NF-kappaB activation characterized by an early activation in the arterial media and a late activation coupled with high levels of inhibitor of IkappaB kinase (IKK) activity in intima. These findings prompted us to elucidate the role for the different phases of NF-kappaB activation and IKK in the progress of vascular repair. Our results show that blockade of the early NF-kappaB activation by perivascular administration of pyrrolidine dithiocarbamate transiently attenuates the expression of proinflammatory genes in the injured vessels but does not affect intimal formation. Interruption of IKKbeta by overexpressing a dominant-negative IKKbeta in the injured artery effectively inhibited the late phase of NF-kappaB activation, resulting in down-regulation of inducible nitric oxide synthase, tumor necrosis factor alpha, and monocyte chemoattractant protein-1 expression in conjunction with a 36% reduction in intima size, albeit with a lack of inhibitory effect on the early NF-kappaB activation. Collectively, these findings show that the IKKbeta-mediated late-phase NF-kappaB activation contributes to intimal hyperplasia and the accompanied vascular inflammatory responses.


Asunto(s)
Quinasa I-kappa B/fisiología , FN-kappa B/fisiología , Transducción de Señal/fisiología , Túnica Íntima/patología , Vasculitis/etiología , Angioplastia/efectos adversos , Animales , Apoptosis , Arterias Carótidas/patología , Proliferación Celular , Hiperplasia , Masculino , Músculo Liso Vascular/patología , Ratas , Ratas Sprague-Dawley
5.
Artículo en Inglés | MEDLINE | ID: mdl-15853755

RESUMEN

Growing evidence suggests that statins are more than simple lipid-lowering drugs. The so called pleiotropic effects of statins include multiple actions on cells of the vasculature. A large number of studies have confirmed that these compounds exert beneficial effects by mechanisms unrelated to cholesterol metabolism. For example, statins have been shown to inhibit the migration and proliferation of vascular smooth muscle cells (VSMC), and to induce apoptosis in this cell type. It is not yet clear if the induction of apoptosis in VSMC by statins is beneficial or detrimental. In the context of post-angioplasty restenosis, recurrent plaque growth after intervention, the inhibition of neointimal proliferation as well as a reduction of neointimal cell numbers by apoptosis is appealing. Multiple animal studies and clinical trials have therefore been undertaken to investigate effects of statin treatment on the development of restenosis, with very controversial results. Conversely, in advanced atherosclerotic lesions VSMC in the intima may stabilize the plaque and prevent plaque rupture by synthesizing collagen. VSMC in media adjacent to plaque areas or restenotic lesions should not be exposed to apoptosis promoting agents. In this context, recent evidence suggests that pravastatin protects such lesions by inhibiting inflammation and macrophage activation Our recent findings together with observations from other groups suggest that neointima cells are more sensitive to the induction of apoptosis than media VSMC. Importantly, statins were found to preferentially induce apoptosis in neointimal VSMC in our study. The purpose of the present review is to summarize statin effects on proliferation and apoptosis in VSMC in vitro and in vivo. Furthermore, the development of drug-coated stents may help to deliver high local doses of statins to enhance their effectiveness in the treatment of post-angioplasty restenosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Reestenosis Coronaria/prevención & control , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Animales , Humanos , Músculo Liso Vascular/patología , Músculo Liso Vascular/fisiología , Miocitos del Músculo Liso/patología , Miocitos del Músculo Liso/fisiología
6.
J Cell Physiol ; 204(3): 881-8, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15795939

RESUMEN

n-3 polyunsaturated fatty acids (PUFAs) have been shown to exert beneficial effects in the prevention of cardiovascular disease, inflammation, and on tumor growth. To investigate effects of PUFAs on proliferation and apoptosis in endothelial cells, we tested the n-3 PUFA docosahexaenoic acid (DHA) and the n-6 PUFA arachidonic acid (AA) in human umbilical vein endothelial cells (HUVEC). The mitochondrial membrane potential (MMP) and the production of reactive oxygen species were examined by flow cytometry. Phosphorylation of p53 or p38 MAP kinase, and total levels of p53 were measured by Western blot. DNA binding activity of p53 was analyzed with a TransAM transcription factor assay kit. Tube formation was assessed on Matrigel. In proliferating HUVEC, but not in confluent cells, DHA reduced cell viability and induced apoptosis, as demonstrated by increases in membrane leakage (propidium iodide (PI) staining), Annexin-V binding, sub G(1) phase in the cell cycle, and TUNEL-positive cells. AA had no effect on these parameters. In addition to a reduced MMP and increased reactive oxygen species, phosphorylation of p38 and p53 (serine 15) and impaired DNA binding of p53 were observed. There was no change in total levels of p53. The p38 inhibitor SB203580 had no effect on Annexin V binding. DHA also attenuated HUVEC tube formation. Taken together, DHA induces apoptosis in proliferating, but not in resting HUVEC, potentially via the phosphorylation of p53, resulting in decreased p53 DNA binding. The results suggest that anti-angiogenic effects of DHA may be due to induction of apoptosis in proliferating endothelial cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Ácidos Docosahexaenoicos/farmacología , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Ácido Araquidónico/farmacología , Proliferación Celular/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , Células Cultivadas , ADN/metabolismo , Fragmentación del ADN/efectos de los fármacos , Células Endoteliales/metabolismo , Activación Enzimática/efectos de los fármacos , Humanos , Potenciales de la Membrana/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Fosfoserina/metabolismo , Fosfotirosina/metabolismo , Superóxidos/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Cordón Umbilical/citología , Cordón Umbilical/efectos de los fármacos , Cordón Umbilical/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
7.
Arterioscler Thromb Vasc Biol ; 25(3): 546-52, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15618544

RESUMEN

OBJECTIVES: Sphingosine 1-phosphate (S1P) is a bioactive phospholipid acting both as a ligand for the G protein-coupled receptors S1P1-5 and as a second messenger. Because S1P1 knockout is lethal in the transgenic mouse, an alternative approach to study the function of S1P1 in endothelial cells is needed. METHODS AND RESULTS: All human endothelial cells analyzed expressed abundant S1P1 transcripts. We permanently silenced (by RNA interference) the expression of S1P1 in the human endothelial cell lines AS-M.5 and ISO-HAS.1. The S1P1 knock-down cells manifested a distinct morphology and showed neither actin ruffles in response to S1P nor an angiogenic reaction. In addition, these cells were more sensitive to oxidant stress-mediated injury. New S1P1-dependent gene targets were identified in human endothelial cells. S1P1 silencing decreased the expression of platelet-endothelial cell adhesion molecule-1 and VE-cadherin and abolished the induction of E-selectin after cell stimulation with lipopolysaccharide or tumor necrosis factor-alpha. Microarray analysis revealed downregulation of further endothelial specific transcripts after S1P1 silencing. CONCLUSIONS: Long-term silencing of S1P1 enabled us for the first time to demonstrate the involvement of S1P1 in key functions of endothelial cells and to identify new S1P1-dependent gene targets.


Asunto(s)
Endotelio Vascular/fisiología , Biología Molecular/métodos , ARN Interferente Pequeño/genética , Receptores de Lisoesfingolípidos/genética , Receptores de Lisoesfingolípidos/metabolismo , Antígenos CD , Cadherinas/genética , Línea Celular , Endotelio Vascular/citología , Silenciador del Gen , Hemangiosarcoma , Humanos , Microcirculación , Neovascularización Fisiológica/fisiología , Análisis de Secuencia por Matrices de Oligonucleótidos , Estrés Oxidativo/fisiología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética , Circulación Pulmonar , ARN Mensajero/genética , Sistemas de Mensajero Secundario/fisiología , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/farmacología , Venas Umbilicales/citología , Neoplasias Vasculares , Vasculitis/genética , Vasculitis/inmunología , Vasculitis/fisiopatología
8.
Blood ; 104(9): 2761-6, 2004 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-15242875

RESUMEN

Endothelial progenitor cells (EPCs) play a role in the repair of ischemic or injured tissue. Because endothelial injury can be associated with apoptosis, we have investigated whether apoptotic bodies from mature endothelial cells (ECs) may affect growth and differentiation of EPCs in vitro. A 24-hour incubation of isolated human EPCs with apoptotic bodies-rich medium (ABRM) from ECs led to a significant increase in the number of spindle-shaped attached cells. EPCs were characterized by DiI-Ac-LDL/lectin staining and measurement of CD34 and kinase insert domain receptor (KDR) expression. The treatment with ABRM resulted in a 2-fold increase of DiI-Ac-LDL/lectin-positive cells and up-regulation of CD34 (22% +/- 2% versus 13% +/- 3%, P < .05 and KDR (49% +/- 12% versus 19% +/- 7%, P < .05). Fluorescence and confocal laser microscopy demonstrated the uptake of apoptotic bodies by the EPCs. Apoptotic bodies-depleted medium had no effect, whereas the incubation with suspension of apoptotic bodies induced effects similar to those of ABRM. Our results suggest that apoptotic bodies from ECs are taken up by EPCs, increasing their number and differentiation state. Such a mechanism may facilitate the repair of injured endothelium and may represent a new signaling pathway between progenitor and damaged somatic cells.


Asunto(s)
Apoptosis , Endotelio Vascular/citología , Células Madre/citología , Adhesión Celular , Diferenciación Celular , Proliferación Celular , Forma de la Célula , Células Cultivadas , Medios de Cultivo Condicionados/química , Citometría de Flujo , Humanos , Microscopía Fluorescente , Fagocitosis , Venas Umbilicales
9.
Circ Res ; 94(6): 776-84, 2004 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-14963004

RESUMEN

Leukocyte recruitment is crucial for the response to vascular injury in spontaneous and accelerated atherosclerosis. Whereas the mechanisms of leukocyte adhesion to endothelium or matrix-bound platelets have been characterized, less is known about the proadhesive role of smooth muscle cells (SMCs) exposed after endothelial denudation. In laminar flow assays, neointimal rat SMCs (niSMCs) supported a 2.5-fold higher arrest of monocytes and "memory" T lymphocytes than medial SMCs, which was dependent on both P-selectin and VLA-4, as demonstrated by blocking antibodies. The increase in monocyte arrest on niSMCs was triggered by the CXC chemokine GRO-alpha and fractalkine, whereas "memory" T cell arrest was mediated by stromal cell-derived factor (SDF)-1alpha. This functional phenotype was paralleled by a constitutively increased mRNA and surface expression of P-selectin and of relevant chemokines in niSMCs, as assessed by real-time PCR and flow cytometry. The increased expression of P-selectin in niSMCs versus medial SMCs was associated with enhanced NF-kappaB activity, as revealed by immunofluorescence staining for nuclear p65 in vitro. Inhibition of NF-kappaB by adenoviral IkappaBalpha in niSMCs resulted in a marked reduction of increased leukocyte arrest in flow. Furthermore, P-selectin expression by niSMCs in vivo was confirmed in a hypercholesterolemic mouse model of vascular injury by double immunofluorescence and by RT-PCR after laser microdissection. In conclusion, we have identified a NF-kappaB-mediated proinflammatory phenotype of niSMCs that is characterized by increased P-selectin and chemokine expression and thereby effectively supports leukocyte recruitment.


Asunto(s)
Quimiotaxis de Leucocito/fisiología , Inflamación/patología , Integrina alfa4beta1/fisiología , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , FN-kappa B/fisiología , Selectina-P/fisiología , Animales , Aorta Torácica/lesiones , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Arteriosclerosis/metabolismo , Arteriosclerosis/patología , Adhesión Celular , Células Cultivadas/efectos de los fármacos , Células Cultivadas/metabolismo , Quimiocina CX3CL1 , Quimiocinas CX3C/farmacología , Constricción Patológica , Citocinas/biosíntesis , Citocinas/genética , Citocinas/fisiología , Endotelio Vascular/lesiones , Regulación de la Expresión Génica , Hipercolesterolemia/metabolismo , Proteínas I-kappa B/fisiología , Inflamación/metabolismo , Integrina alfa4beta1/biosíntesis , Integrina alfa4beta1/genética , Proteínas de la Membrana/farmacología , Ratones , Ratones Noqueados , Monocitos/citología , Músculo Liso Vascular/citología , Inhibidor NF-kappaB alfa , Selectina-P/biosíntesis , Selectina-P/genética , Ratas , Ratas Sprague-Dawley , Receptores de Interleucina-8B/fisiología , Proteínas Recombinantes de Fusión/fisiología , Recurrencia , Reología , Subgrupos de Linfocitos T/citología
10.
Eur J Immunol ; 34(1): 241-50, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14971050

RESUMEN

Cyclopentenone prostaglandins (CP-PG), such as prostaglandin A1 (PGA1) or 15-deoxy-Delta(12,14)-prostaglandin J2 (PGJ2), induce apoptosis in different cell types. PGJ2 is also a potent activator of the peroxisome proliferator-activated receptor-gamma (PPARgamma). We investigated whether PPARgamma regulates CP-PG-induced apoptosis in endothelial cells (EC). We show that CP-PG induce apoptosis in human umbilical vein EC (HUVEC). Incubation with PGA1 or PGJ2 for 24 h reduced HUVEC number and viability, while the synthetic activators Wy14643 or rosiglitazone had no effect. Flow cytometry and cell cycle analysis revealed externalized phosphatidylserine, caspase-3 activation, and an increased percentage of cells with a reduced DNA content by CP-PG treatment. EMSA demonstrated an activation of PPARgamma by PGJ2 and rosiglitazone. Immunohistochemistry of HUVEC and immunoblot analyses of protein extracts showed that PPARgamma was localized in the nuclei of HUVEC, and that CP-PG treatment decreased the amount of PPARgamma protein. This degradation was prevented by a pan-caspase inhibitor. Treatment of differentiated, endothelial-like PPARgamma-deficient stem cells, or of HUVEC transfected with dominant-negative PPARgamma with CP-PG, induced cell death and apoptosis. Our findings show that PGA1 and PGJ2 induce apoptosis in endothelial cells independent of PPARgamma. As the synthesis of PGJ2 is increased at sites of inflammation, our results may suggest a possible mechanism for endothelial damage.


Asunto(s)
Apoptosis/efectos de los fármacos , Prostaglandinas A/farmacología , Venas Umbilicales/efectos de los fármacos , Caspasa 3 , Caspasas/efectos de los fármacos , Endotelio/efectos de los fármacos , Endotelio/metabolismo , Humanos , Receptores Citoplasmáticos y Nucleares/metabolismo , Factores de Transcripción/metabolismo
11.
Blood ; 103(7): 2585-92, 2004 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-14645014

RESUMEN

Despite the fact that lysophosphatidic acid (LPA) has been identified as a main platelet-activating lipid of mildly oxidized low-density lipoprotein (LDL) and human atherosclerotic lesions, it remains unknown whether it is capable of activating platelets in blood. We found that LPA at concentrations slightly above plasma levels induces platelet shape change, aggregation, and platelet-monocyte aggregate formation in blood. 1-alkyl-LPA (16:0 fatty acid) was almost 20-fold more potent than 1-acyl-LPA (16:0). LPA directly induced platelet shape change in blood and platelet-rich plasma obtained from all blood donors. However, LPA-stimulated platelet aggregation in blood was donor dependent. It could be completely blocked by apyrase and antagonists of the platelet adenosine diphosphate (ADP) receptors P2Y1 and P2Y12. These substances also inhibited LPA-induced aggregation of platelet-rich plasma and aggregation and serotonin secretion of washed platelets. These results indicate a central role for ADP-mediated P2Y1 and P2Y12 receptor activation in supporting LPA-induced platelet aggregation. Platelet aggregation and platelet-monocyte aggregate formation stimulated by LPA was insensitive to inhibition by aspirin. We conclude that LPA at concentrations approaching those found in vivo can induce platelet shape change, aggregation, and platelet-monocyte aggregate formation in whole blood and suggest that antagonists of platelet P2Y1 and P2Y12 receptors might be useful preventing LPA-elicited thrombus formation in patients with cardiovascular diseases.


Asunto(s)
Arteriopatías Oclusivas/sangre , Lisofosfolípidos/farmacología , Lisofosfolípidos/fisiología , Proteínas de la Membrana/fisiología , Activación Plaquetaria/fisiología , Receptores Purinérgicos P2/fisiología , Adenosina Difosfato/farmacología , Plaquetas/efectos de los fármacos , Plaquetas/fisiología , Tamaño de la Célula/efectos de los fármacos , Humanos , Cinética , Activación Plaquetaria/efectos de los fármacos , Receptores Purinérgicos P2Y1 , Receptores Purinérgicos P2Y12 , Transducción de Señal
12.
Atherosclerosis ; 169(2): 251-8, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12921976

RESUMEN

In the context of atherogenesis and restenosis, vascular smooth muscle cell (SMC) proliferation and apoptosis play a crucial role. Inhibitors of 3-hydroxy-3-methylglutaryl coenzyme A reductase (statins) have been shown to inhibit the migration and proliferation of SMC, and to induce apoptosis in different cell types including SMC. However, it is not known whether these agents induce apoptosis in neointimal SMC. We investigated the effects of statin treatment on neointimal SMC as compared to medial cells by using trypan blue counting, MTT test, Annexin V staining, cell cycle analysis and a co-culture model. The incubation of neointimal or medial SMC with lovastatin reduced the MTT activity as well as the total cell number, and increased the amount of trypan blue positive cells, indicative of cell death. We tested by staining with Annexin V/propidium iodide, specific antibodies to active caspase-3, TUNEL reaction, and by the appearance of a sub-G1 peak, whether the observed increase in cell death was due to apoptosis. After treatment with lovastatin, programmed cell death was slightly increased in medial SMC, while neointimal cells showed a pronounced rate of apoptosis. In an attempt to mimic early phases of restenosis in vitro by seeding low density neointimal cells onto high density medial cells, we found that statin treatment induced cell death preferentially in the neointimal SMC. Our results suggest that statins enhance the rate of apoptosis in neointimal SMC, which may be an interesting feature to reduce restenosis after successful angioplasty.


Asunto(s)
Apoptosis/efectos de los fármacos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Músculo Liso Vascular/efectos de los fármacos , Animales , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Etiquetado Corte-Fin in Situ , Lovastatina/farmacología , Masculino , Células Musculares/citología , Células Musculares/efectos de los fármacos , Músculo Liso Vascular/citología , Ratas , Ratas Sprague-Dawley , Túnica Íntima/citología
13.
Biochem Biophys Res Commun ; 307(2): 322-6, 2003 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-12859958

RESUMEN

Prostaglandins are a family of structurally related molecules formed by many cells in response to extrinsic stimuli. A member of this family, 15-deoxy-Delta(12,14)-PGJ(2) (15d-PGJ(2)), shows unique biological properties including anti-inflammatory, anti-viral, and anti-tumour activity, and has attracted much attention as a high affinity ligand for the peroxisome proliferator-activated receptor gamma. Increasing evidence points to additional effects. We investigated several structurally related prostaglandins in comparison to 15d-PGJ(2) with respect to their apoptosis-inducing capacity in human umbilical endothelial cells (HUVEC). Cell viability was tested with a modified MTT assay and apoptosis was detected by Annexin V staining and cell cycle analysis by flow cytometry. Incubation of confluent HUVECs with 15d-PGJ(2) markedly reduced endothelial cell viability which was due to apoptosis. In contrast, none of the other PGs tested affected cell viability. Interestingly, the cyclopentenone ring alone dose-dependently reduced cell viability and significantly induced apoptosis in HUVECs with as low a concentration as 0.25 microM. In conclusion, we report that the cyclopentenone moiety of cyPGs is an essential component for the apoptosis-inducing properties of 15d-PGJ(2). For 15d-PGJ(2) the position of the cyclopentenone ring in conjunction with the side chains yields a molecule with unique biological properties.


Asunto(s)
Apoptosis , Ciclopentanos/química , Ciclopentanos/farmacología , Endotelio Vascular/citología , Prostaglandinas/química , Prostaglandinas/farmacología , Línea Celular , Humanos , Estructura Molecular , Prostaglandina D2/análogos & derivados , Prostaglandina D2/química , Prostaglandina D2/farmacología
14.
Trends Cardiovasc Med ; 13(5): 201-6, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12837583

RESUMEN

Bone marrow of adults contains a subtype of progenitor cells that have the capacity to differentiate into mature endothelial cells and have therefore been termed endothelial progenitor cells (EPCs). Of the three cell markers (CD133, CD34, and the vascular endothelial growth factor receptor 2) that characterize the early functional EPCs, located predominantly in the bone marrow, EPCs obviously lose CD133/CD34 and start to express CD31, vascular endothelial cadherin, and von Willebrand factor when migrating to the circulation. Various isolation procedures of EPCs from different sources by using adherence culture or magnetic microbeads have been described, but published findings with regard to the number of EPCs in the peripheral circulation of healthy adults are scanty and no data regarding the lifetime of EPCs in vivo exist. Clinical studies employing EPCs for neovascularization of ischemic organs have just been started; however, the mechanisms stimulating or inhibiting the differentiation of bone marrow-derived EPCs in vivo and the signals causing their adhesion, migration, and homing to sites of injured tissue are largely unknown at present.


Asunto(s)
Endotelio/citología , Neovascularización Fisiológica , Células Madre/citología , Separación Celular , Humanos
15.
FASEB J ; 17(9): 1099-101, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12692090

RESUMEN

The cyclopentenone prostaglandins (cPG) 15-deoxy-Delta12,14-prostaglandin J2 (dPGJ2) and PGA1 can inhibit multiple steps in nuclear factor (NF)-kappaB signaling and can induce cell death. Here we characterized the effects of dPGJ2 and PGA1 on the inflammatory induction of endothelial cell adhesion molecules (CAM). Pretreatment of endothelial cells with dPGJ2 or PGA1 at low concentrations dose dependently inhibited the up-regulation of CAM expression and monocyte arrest by tumor necrosis factor (TNF)-alpha but not expression of inhibitor of apoptosis proteins. Only at high concentrations, cPG enhanced TNF-alpha-induced cell death and inhibited TNF-alpha-induced IkappaB-alpha kinase (IKK) activation, IkappaB-alpha degradation, and NF-kappaB/p65 translocation, while promoting AP-1/c-jun phosphorylation. Expression of an IKK-beta mutant (C179A) resistant to interaction with cPG impaired cell death induction but not inhibition of CAM up-regulation by cPG. Gel shift and reporter gene analysis revealed that cPG at low concentrations directly impaired DNA binding of NF-kappaB and NF-kappaB-dependent transactivation. The synthetic analogs dPGA1 or dPGA2 were ineffective, indicating structural specificity of cPG. Thus, the suppression of endothelial CAM up-regulation with cPG is dissociated from cell death sensitization and IKK inhibition above threshold concentrations and related to interference with NF-kappaB binding. Our findings define distinct mechanisms for anti-inflammatory and proapoptotic effects of cPG in endothelial cells.


Asunto(s)
Moléculas de Adhesión Celular/biosíntesis , Endotelio Vascular/metabolismo , Prostaglandina D2/farmacología , Prostaglandinas A/farmacología , Proteínas Serina-Treonina Quinasas/metabolismo , Apoptosis , Relación Dosis-Respuesta a Droga , Endotelio Vascular/efectos de los fármacos , Humanos , Quinasa I-kappa B , Proteínas I-kappa B/metabolismo , Molécula 1 de Adhesión Intercelular/biosíntesis , Modelos Biológicos , Inhibidor NF-kappaB alfa , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Prostaglandina D2/análogos & derivados , Factor de Transcripción ReIA , Factor de Necrosis Tumoral alfa/farmacología , Regulación hacia Arriba , Molécula 1 de Adhesión Celular Vascular/biosíntesis
16.
Arterioscler Thromb Vasc Biol ; 23(7): 1185-9, 2003 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-12714439

RESUMEN

Postnatal bone marrow contains a subtype of progenitor cells that have the capacity to migrate to the peripheral circulation and to differentiate into mature endothelial cells. Therefore, these cells have been termed endothelial progenitor cells (EPCs). The isolation of EPCs by adherence culture or magnetic microbeads has been described. In general, EPCs are characterized by the expression of 3 markers, CD133, CD34, and the vascular endothelial growth factor receptor-2. During differentiation, EPCs obviously lose CD133 and start to express CD31, vascular endothelial cadherin, and von Willebrand factor. EPCs seem to participate in endothelial repair and neovascularization of ischemic organs. Clinical studies using EPCs for neovascularization have just been started; however, the mechanisms stimulating or inhibiting the differentiation of EPC in vivo and the signals causing their migration and homing to sites of injured endothelium or extravascular tissue are largely unknown at present. Thus, future studies will help to explore areas of potential basic research and clinical application of EPCs.


Asunto(s)
Diferenciación Celular/fisiología , Movimiento Celular/fisiología , Células Endoteliales/fisiología , Células Madre Mesenquimatosas/fisiología , Antígeno AC133 , Angioplastia de Balón , Antígenos CD , Antígenos CD34/metabolismo , Recuento de Células , Técnicas de Cultivo de Célula , Células Endoteliales/trasplante , Endotelio Vascular/lesiones , Endotelio Vascular/fisiología , Glicoproteínas/metabolismo , Humanos , Péptidos/metabolismo , Regeneración , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Factores de Crecimiento Endotelial Vascular/metabolismo
17.
Clin Cancer Res ; 8(3): 863-9, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11895920

RESUMEN

Alpha-tocopheryl succinate (alpha-TOS), a redox-inactive analogue of vitamin E, is a strong inducer of apoptosis, whereas alpha-tocopherol (alpha-TOH) lacks apoptogenic activity (J. Neuzil et al., FASEB J., 15: 403-415, 2001). Here we investigated the possible antineoplastic activities of alpha-TOH and alpha-TOS and further explored the potential of alpha-TOS as an antitumor agent. Using nude mice with colon cancer xenografts, we found that alpha-TOH exerted modest antitumor activity and acted by inhibiting tumor cell proliferation. In contrast, alpha-TOS showed a more profound antitumor effect, at both the level of inhibition of proliferation and induction of tumor cell apoptosis. alpha-TOS was nontoxic to normal cells and tissues, triggered apoptosis in p53(-/-) and p21(Waf1/Cip1(-/-)) cancer cells, and exerted a cooperative proapoptotic activity with tumor necrosis factor-related apoptosis-inducing ligand (Apo2 ligand) due to differences in proapoptotic signaling. Finally, alpha-TOS cooperated with tumor necrosis factor-related apoptosis-inducing ligand in suppression of tumor growth in vivo. Vitamin E succinate is thus a potent and highly specific anticancer agent and/or adjuvant of considerable therapeutic potential.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Neoplasias del Colon/tratamiento farmacológico , Glicoproteínas de Membrana/metabolismo , Células Tumorales Cultivadas/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo , Vitamina E/análogos & derivados , Vitamina E/farmacología , Animales , Proteínas Reguladoras de la Apoptosis , Caspasas/metabolismo , Ensayo de Unidades Formadoras de Colonias , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/metabolismo , Sinergismo Farmacológico , Humanos , Ratones , Contracción Miocárdica/efectos de los fármacos , Contracción Miocárdica/fisiología , Miocardio/citología , Invasividad Neoplásica , Ligando Inductor de Apoptosis Relacionado con TNF , Tocoferoles , Transfección , Proteína p53 Supresora de Tumor/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , alfa-Tocoferol/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA