Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Hypertension ; 56(2): 247-52, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20530298

RESUMEN

Calcium is an essential signaling molecule that controls vascular smooth muscle cell (VSMC) contraction, proliferation, and differentiation. Here, we show that the calcium antagonist nifedipine inhibits VSMC dedifferentiation in vitro and in vivo. Differentiated VSMCs cultured on laminin-coated dishes were transferred to laminin-free dishes to induce dedifferentiation. Induction of dedifferentiation resulted in the upregulation of nonmuscle myosin heavy chain expression, a marker of dedifferentiation, and the downregulation of smooth muscle myosin heavy chain expression, a marker of differentiation. Nifedipine significantly inhibited both the induction of these phenotypic changes and upregulation of Akt signaling in these cells. Administration of nifedipine at a low concentration that did not affect blood pressure could inhibit the increase in nonmuscle myosin heavy chain expression and decrease in smooth muscle myosin heavy chain expression in a rat balloon-injury model. Furthermore, nifedipine suppressed neointimal hyperplasia and upregulation of Akt signaling. However, phospho-Akt expression was not suppressed in the regenerating arterial endothelium of the nifedipine-treated rats. The inhibitory effect of the downregulation of Akt signaling by dominant-negative Akt on the induction of VSMC dedifferentiation in the intima was identical to that of nifedipine. In contrast, upregulation of Akt signaling by transfection of the cells with a constitutively active Akt reversed the nifedipine-induced inhibition of VSMC dedifferentiation. In conclusion, nifedipine inhibits VSMC dedifferentiation by suppressing Akt signaling, thereby preventing neointimal thickening.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Músculo Liso Vascular/citología , Músculo Liso Vascular/fisiología , Nifedipino/farmacología , Proteínas Proto-Oncogénicas c-akt/fisiología , Adenoviridae/genética , Adenoviridae/fisiología , Animales , Aorta/citología , Aorta/efectos de los fármacos , Aorta/fisiología , Aorta/virología , Regulación hacia Abajo/efectos de los fármacos , Humanos , Masculino , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/virología , Proteínas Proto-Oncogénicas c-akt/genética , Ratas , Ratas Sprague-Dawley , Túnica Íntima/efectos de los fármacos , Túnica Íntima/fisiología , Regulación hacia Arriba/efectos de los fármacos , Vasodilatadores/farmacología
2.
Hypertension ; 43(6): 1208-13, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15117910

RESUMEN

Hyperhomocysteinemia is an independent risk factor for the development of atherosclerosis. However, the underlying mechanism of endothelial cell injury in hyperhomocysteinemia has not been elucidated. In this study, we examined the effect of homocysteine (Hcy) on Fas-mediated apoptosis in endothelial cells. Hcy-induced upregulation of Fas in endothelial cells (ECs) in a dose-dependent manner. At the same time, Hcy increased intracellular peroxide in ECs. Hcy-induced Fas expression was inhibited by the treatment with catalase. Hcy increased NF-kappaB DNA binding activity, and adenovirus-mediated transfection of a Ikappa-B mutant (Ikappa-B mt) gene inhibited Hcy-induced Fas expression. ECs were sensitive to Fas-mediated apoptosis when exposed to Hcy. Under these condition, Ikappa-B mt protected ECs from Fas-mediated apoptosis. In addition, Hcy inhibited expression of the caspase-8 inhibitor FLICE-inhibitory protein (FLIP). Adenovirus-mediated transfection of constitutively active Akt gene abolished the Hcy-mediated downregulation of FLIP. These data suggest that upregulation of Fas expression and downregulation of FLIP is a mechanism through which Hcy induces EC apoptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Homocisteína/toxicidad , Péptidos y Proteínas de Señalización Intracelular , Receptor fas/fisiología , Animales , Arteriosclerosis/etiología , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD , Proteínas Portadoras/fisiología , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Proteínas I-kappa B/genética , Proteínas I-kappa B/fisiología , Ratones , FN-kappa B/fisiología , Estrés Oxidativo , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/fisiología , Proteínas Proto-Oncogénicas c-akt , Proteínas Recombinantes de Fusión/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Venas Umbilicales/citología , Regulación hacia Arriba/efectos de los fármacos , Receptor fas/biosíntesis , Receptor fas/genética
3.
Hypertension ; 43(4): 880-4, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-14967838

RESUMEN

A growing body of evidence has shown that Fas, a death receptor, mediates apoptosis-unrelated biological effects. Here, we report that Fas engagement with Fas ligand induced activation of Akt and upregulation of endothelial nitric oxide synthase expression without induction of apoptosis. In the presence of the phosphatidylinositol 3-kinase inhibitor wortmannin, Fas ligand, however, induced apoptosis instead of upregulation of endothelial nitric oxide synthase expression. In vivo, systolic blood pressure was slightly higher in mutant mice with decreased cell surface Fas expression (lpr mice) compared with wild-type mice. In addition, chronic inhibition of nitric oxide synthesis by N(G)-nitro-l-arginine induced a progressive increase in the levels of blood pressure in wild-type mice, whereas no further increase in the levels of blood pressure was observed in lpr mice. Furthermore, acetylcholine caused a lesser endothelium-dependent relaxation of the strips from lpr mice compared with wild-type mice, although the vasoconstrictor potency of phenylephrine was not different between the two groups. These findings indicate that Fas signaling may have a role in the regulation of endothelial function and blood pressure through modulating endothelial nitric oxide synthase expression in the Akt signal-dependent manner.


Asunto(s)
Endotelio Vascular/enzimología , Glicoproteínas de Membrana/fisiología , Óxido Nítrico Sintasa/biosíntesis , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Proto-Oncogénicas/fisiología , Transducción de Señal , Receptor fas/fisiología , Acetilcolina/farmacología , Androstadienos/farmacología , Animales , Apoptosis/efectos de los fármacos , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/fisiología , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/enzimología , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Inducción Enzimática/fisiología , Inhibidores Enzimáticos/farmacología , Proteína Ligando Fas , Humanos , Insulina/farmacología , Células Jurkat/efectos de los fármacos , Células Jurkat/metabolismo , Masculino , Ratones , Ratones Endogámicos MRL lpr , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa de Tipo II , Óxido Nítrico Sintasa de Tipo III , Nitroarginina/farmacología , Fenilefrina/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt , Proteínas Recombinantes/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Venas Umbilicales , Vasoconstrictores/farmacología , Wortmanina
4.
Hypertens Res ; 26(6): 503-8, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12862208

RESUMEN

Calcium antagonists normalize endothelial dysfunction and improve the clinical outcome in patients with hypertension. However, the mechanism underlying these beneficial effects remains to be elucidated. Here, we show that the calcium antagonist nifedipine upregulates the expression of manganese superoxide dismutase (Mn SOD), an endogenous antioxidant enzyme, in vascular smooth muscle cells (VSMC) via cellular interactions between VSMC and endothelial cells (EC). Nifedipine induced upregulation of Mn SOD activity and expression in VSMC when cocultured with EC but not when cultured individually. NG-Monomethyl-L-arginine (L-NMMA), an inhibitor of nitric oxide (NO) synthesis, inhibited the upregulation of Mn SOD expression induced by nifedipine. Additionally, N-ethyl-2-(1-ethyl-2-hydroxy-2-nitrosohydrazino) ethanamine, a NO donor, reversed this inhibition by L-NMMA, indicating that NO may be involved in the mechanism underlying the nifedipine-induced upregulation of Mn SOD in VSMC. Preincubation of VSMC with Mn SOD antisense oligodeoxyribonucleotides (ODN) blocked the suppressive effects of nifedipine on DNA synthesis in VSMC cocultured with EC, whereas sense ODN had no effect. We conclude that the calcium antagonist nifedipine induces upregulation of Mn SOD expression in VSMC via NO derived from EC. This finding may provide some insight into the mechanism underlying the beneficial effects of calcium antagonists in patients with hypertension.


Asunto(s)
Bloqueadores de los Canales de Calcio/farmacología , Células Endoteliales/fisiología , Músculo Liso Vascular/enzimología , Nifedipino/farmacología , Superóxido Dismutasa/biosíntesis , Western Blotting , División Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Células Endoteliales/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Humanos , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Óxido Nítrico/fisiología , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo III , Oligonucleótidos Antisentido/farmacología , Superóxido Dismutasa/genética , Tubulina (Proteína)/biosíntesis , Regulación hacia Arriba/efectos de los fármacos , omega-N-Metilarginina/farmacología
5.
Hypertension ; 42(3): 342-8, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12874095

RESUMEN

Dietary supplementation with eicosapentaenoic acid (EPA) improves the prognosis of chronic inflammatory diseases, including atherosclerosis. The mechanism underlying these beneficial effects, however, remains to be elucidated. Here we show that EPA protects endothelial cells from anoikis through upregulation of the cellular FLICE (Fas-associating protein with death domain-like interleukin-1-converting enzyme)-inhibitory protein (cFLIP), an endogenous inhibitor of caspase-8. EPA-induced upregulation of cFLIP expression was partially suppressed by the phosphatidylinositol-3-kinase inhibitor wortmannin. Conversely, treatment with insulinlike growth factor-1 (IGF-1), an activator of phosphatidylinositol-3-kinase/Akt signaling, or infection with an adenoviral construct expressing the constitutively active Akt gene induced upregulation of cFLIP expression. In addition, pretreatment of endothelial cells with either EPA or IGF-1 protected them from anoikis, suggesting that EPA-induced protection against anoikis is partially mediated through activation of Akt. On the other hand, when endothelial cells were already detached, treatment of these cells with EPA but not with IGF-1 protected them against anoikis. Importantly, EPA restored cFLIP expression without activating Akt signaling in detached endothelial cells, whereas IGF-1 had no effect. Additionally, exogenously restored expression of cFLIP by the tetracycline-regulated adenovirus system protected endothelial cells against anoikis. Furthermore, EPA was protective against the loss of endothelium in an organ culture of rat aortas. These findings suggest that EPA protects against endothelial cell anoikis through restoration of cFLIP expression, which might contribute to the mechanism underlying the beneficial effects of EPA in patients with hypertension.


Asunto(s)
Anoicis/efectos de los fármacos , Proteínas Portadoras/metabolismo , Ácido Eicosapentaenoico/farmacología , Endotelio Vascular/efectos de los fármacos , Péptidos y Proteínas de Señalización Intracelular , Proteínas Serina-Treonina Quinasas , Aorta/citología , Aorta/efectos de los fármacos , Apoptosis/efectos de los fármacos , Western Blotting , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD , Proteínas Portadoras/genética , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Expresión Génica , Vectores Genéticos/genética , Humanos , Factor I del Crecimiento Similar a la Insulina/farmacología , Técnicas de Cultivo de Órganos , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Transfección
6.
J Biol Chem ; 278(17): 15185-91, 2003 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-12576484

RESUMEN

Fas ligand (FasL) is a member of tumor necrosis factor family that induces apoptosis in target cells that express Fas. The function of FasL during inflammation remains controversial. In this study, we examined the role of vascular endothelial FasL during acute myocardial ischemia-reperfusion that is closely associated with inflammation. Transgenic mouse lines were established that overexpress human FasL on endothelium under the control of the vascular endothelial cadherin promoter. Expression of FasL transgene was detected at both mRNA and protein levels, and functional transgene-encoded FasL protein was specifically expressed on the surface of vascular endothelial cells. Transgenic mice developed normally and had normal hearts. When subjected to 30 min of myocardial ischemia and 72 h of reperfusion, myocardial infarct size was reduced by 42% in the transgenic mice compared with nontransgenic littermates (p < 0.05). Moreover, hemodynamic data demonstrated that transgenic hearts performed better following ischemia and reperfusion compared with nontransgenic hearts. Myocardial neutrophil infiltration was reduced by 54% after 6 h of reperfusion in transgenic hearts (p < 0.01). Neutrophil depletion prior to ischemia-reperfusion injury led to smaller infarcts that were not different between transgenic and nontransgenic mice, suggesting that endothelial FasL may attenuate ischemia-reperfusion injury by abating the inflammatory response. These results indicate that vascular endothelial FasL may exert potent anti-inflammatory actions in the setting of myocardial ischemia-reperfusion injury.


Asunto(s)
Endotelio Vascular/metabolismo , Glicoproteínas de Membrana/fisiología , Daño por Reperfusión Miocárdica/prevención & control , Animales , Cadherinas/genética , Línea Celular , Quimiotaxis de Leucocito , Proteína Ligando Fas , Corazón/fisiopatología , Hemodinámica , Glicoproteínas de Membrana/biosíntesis , Glicoproteínas de Membrana/genética , Ratones , Ratones Transgénicos , Isquemia Miocárdica/patología , Daño por Reperfusión Miocárdica/patología , Neutrófilos , Tamaño de los Órganos , Regiones Promotoras Genéticas
7.
Neurobiol Aging ; 24(3): 437-51, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12600720

RESUMEN

The application of beta-amyloid (Abeta) is cytotoxic to endothelial cells, promotes vasoconstriction and impairs nitric oxide (NO) generation or action. However, there is no information on the effect of intracellular Abeta on endothelial cell biology, although recent studies indicate that neuronal Abeta drives Alzheimer's disease pathogenesis. Since the serine-threonine kinase Akt is crucial to both neuronal and endothelial cell survival as well as eNOS activation, we investigated the effects of Abeta expression on Akt-signaling in cultured endothelial cells. Virally-encoded Abeta42 was proapoptotic and inhibitory to Akt phosphorylation in human umbilical vein endothelial cells (HUVECs). Toxicity was characterized by mitochondrial dysfunction, DNA condensation and activation of caspase-3. Substrates downstream of Akt action, GSK-3beta and eNOS, are underphosphorylated in the presence of Abeta. Constitutive activation of Akt reversed Abeta-induced toxicity and stimulated caspase-3 activity, suggesting that inhibition of Akt signaling is functionally significant. These Abeta effects were mediated, in part, through the derepression of GSK-3beta activation and correlated with reductions in NO production. We conclude that intracellular production of Abeta42 is cytotoxic to endothelial cells and that disruption of the Akt/GSK-3beta cell signaling pathway is involved.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Apoptosis/fisiología , Endotelio Vascular/metabolismo , Glucógeno Sintasa Quinasas/metabolismo , Óxido Nítrico Sintasa/antagonistas & inhibidores , Fragmentos de Péptidos/toxicidad , Proteínas Tirosina Quinasas/fisiología , Sistemas de Mensajero Secundario/fisiología , Transducción de Señal , Agammaglobulinemia Tirosina Quinasa , Caspasa 3 , Caspasas/efectos de los fármacos , Caspasas/metabolismo , Células Cultivadas , Endotelio Vascular/citología , Humanos , Líquido Intracelular , Fosforilación/efectos de los fármacos , Transducción de Señal/fisiología , Transfección , Transgenes/genética , Venas Umbilicales
8.
J Biol Chem ; 277(44): 41888-96, 2002 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-12167628

RESUMEN

Glycogen synthase kinase-3beta (GSK3beta) plays important roles in metabolism, embryonic development, and tumorigenesis. Here we investigated the role of GSK3beta signaling in vascular biology by examining its function in endothelial cells (ECs). In EC, the regulatory phosphorylation of GSK3beta was found to be under the control of phosphoinositide 3-kinase-, MAPK-, and protein kinase A-dependent signaling pathways. The transduction of a nonphosphorylatable constitutively active mutant of GSKbeta promoted apoptosis under the conditions of prolonged serum deprivation or the disruption of cell-matrix attachments. Conversely, the transduction of catalytically inactive GSK3beta promoted EC survival under the conditions of cellular stress. Under normal cell culture conditions, the activation of GSK3beta signaling inhibited the migration of EC to vascular endothelial growth factor or basic fibroblast growth factor. Angiogenesis was inhibited by GSK3beta activation in an in vivo Matrigel plug assay, whereas the inhibition of GSK3beta signaling enhanced capillary formation. These data suggest that GSK3beta functions at the nodal point of converging signaling pathways in EC to regulate vessel growth through its control of vascular cell migration and survival.


Asunto(s)
Glucógeno Sintasa Quinasa 3/fisiología , Músculo Liso Vascular/citología , Neovascularización Fisiológica/fisiología , Proteínas Serina-Treonina Quinasas , Anoicis , Apoptosis , Movimiento Celular , Supervivencia Celular , Células Cultivadas , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Glucógeno Sintasa Quinasa 3 beta , Humanos , Proteínas Quinasas Activadas por Mitógenos/fisiología , Óxido Nítrico/biosíntesis , Fosforilación , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt
9.
Circulation ; 106(3): 356-61, 2002 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-12119253

RESUMEN

BACKGROUND: Calcium antagonists normalize endothelial dysfunction in many cardiovascular diseases. There is no known receptor, however, for calcium antagonists in endothelial cells (ECs). We hypothesized that vascular smooth muscle cells (VSMCs) are involved in the mechanism underlying the normalization of endothelial dysfunction by calcium antagonists. METHODS AND RESULTS: Coculture studies with ECs and VSMCs were performed to determine whether VSMCs mediate modulation of endothelial superoxide dismutase (SOD) activity and expression induced by the calcium antagonist nifedipine. Nifedipine induced upregulation of SOD activity in rat aortic segments but had no effect on SOD expression or activity in ECs or VSMCs cultured individually. When ECs were cocultured with VSMCs, however, nifedipine upregulated SOD expression and activity in ECs. Nifedipine stimulated vascular endothelial growth factor (VEGF) production from VSMCs, and this stimulation of VEGF production was abolished by HOE-140, an antagonist of the bradykinin B(2) receptor. A neutralizing antibody against VEGF inhibited the upregulation of endothelial SOD by nifedipine. In addition, recombinant VEGF induced an increase in the levels of SOD expression in ECs, and supernatant derived from nifedipine-treated VSMCs enhanced NO production from ECs. This increase in NO production by the supernatant was inhibited by preincubation of ECs with SOD antisense oligodeoxyribonucleotides. CONCLUSIONS: The calcium antagonist nifedipine indirectly upregulates endothelial SOD expression by stimulating VEGF production from adjacent VSMCs. This finding may provide further insight into the mechanism underlying the beneficial effects of calcium antagonists in cardiovascular diseases.


Asunto(s)
Bloqueadores de los Canales de Calcio/farmacología , Endotelio Vascular/enzimología , Músculo Liso Vascular/fisiología , Nifedipino/farmacología , Superóxido Dismutasa/biosíntesis , Animales , Aorta/citología , Aorta/enzimología , Comunicación Celular , Células Cultivadas , Técnicas de Cocultivo , Factores de Crecimiento Endotelial/fisiología , Endotelio Vascular/efectos de los fármacos , Humanos , Linfocinas/fisiología , Masculino , Óxido Nítrico/biosíntesis , Ratas , Ratas Wistar , Receptores de Bradiquinina/metabolismo , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
10.
J Am Coll Cardiol ; 39(4): 585-90, 2002 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-11849855

RESUMEN

OBJECTIVES: To examine whether the Fas/Fas ligand system is involved in the pathogenesis of acute myocardial infarction (AMI), we measured the levels of the soluble form of the Fas ligand (sFasL) in the plasma of patients with AMI and stable or unstable angina pectoris (AP). BACKGROUND: The Fas ligand (FasL) is rapidly cleaved off by a metalloproteinase from the cell membrane to become a soluble form as a cytokine. Fas is expressed in most cells, including cardiomyocytes, whereas FasL is mainly expressed in inflammatory cells such as macrophages, which are greatly accumulated in unstable plaque. METHODS: Thirty patients with AMI, 10 patients with unstable AP, 10 patients with stable AP and 30 control subjects were enrolled in the present study. RESULTS: Plasma sFasL levels were significantly elevated on hospital admission in patients with AMI and unstable AP, compared with control subjects. Time-course studies revealed that plasma sFasL levels rapidly decreased within 3 h and then increased again after percutaneous transluminal coronary angioplasty in patients with AMI, but not in patients with stable AP. Importantly, the sFasL levels were higher in the coronary sinus than in the circulation. In addition, in vitro studies showed that the expression of FasL messenger ribonucleic acid was upregulated in mononuclear cells isolated from patients with AMI and that hypoxia stimulated the release of sFasL from isolated mononuclear cells. CONCLUSIONS: This demonstration of elevated levels of sFasL in patients with AMI and unstable AP suggests that activation of the Fas/FasL system may play a pathogenic role in AMI and acute coronary syndromes.


Asunto(s)
Angina de Pecho/sangre , Angina de Pecho/fisiopatología , Angina Inestable/sangre , Angina Inestable/fisiopatología , Glicoproteínas de Membrana/sangre , Infarto del Miocardio/sangre , Infarto del Miocardio/fisiopatología , Anciano , Angina de Pecho/diagnóstico por imagen , Angina Inestable/diagnóstico por imagen , Angiografía Coronaria , Proteína Ligando Fas , Femenino , Hemodinámica/fisiología , Humanos , Masculino , Persona de Mediana Edad , Infarto del Miocardio/diagnóstico por imagen , Índice de Severidad de la Enfermedad , Solubilidad , Factores de Tiempo
11.
Mol Cell Biol ; 22(2): 680-91, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11756562

RESUMEN

Fas and Fas ligand (FasL) expression has been detected in chronic vascular lesions, and Fas-mediated apoptosis of vascular smooth muscle cells (VSMC) may influence the integrity of the atherosclerotic plaque. Here we report that FasL is not expressed by normal VSMC, but its expression is upregulated by stresses that induce apoptosis, including serum deprivation, exposure to the phosphatidylinositol 3-kinase (PI 3-kinase) inhibitor wortmannin, and ablation of Akt signaling. Conversely, constitutive activation of Akt signaling diminished FasL expression in VSMC cultures exposed to low-mitogen media or wortmannin. Under conditions of suppressed PI 3-kinase/Akt signaling, VSMC apoptosis was partially inhibited by treatment with neutralizing antibody against FasL. Suppression of Akt signaling increased the activity of c-Jun N-terminal kinase, and transduction of dominant-negative c-Jun inhibited FasL induction under these conditions. Diminished Akt signaling promoted the cleavage of caspase 3, and both caspase 3 cleavage and FasL induction were inhibited by transduction of dominant-negative caspase 9 or the caspase 8 inhibitor CrmA. Similarly, induction of FasL by the Akt-regulated forkhead transcription factor FKHRL1 was dependent upon caspase and c-Jun activation. Taken together, these results indicate that the sequential activation of caspase 3 and c-Jun participates in the induction of FasL under conditions of suppressed Akt signaling or FKHRL1 activation and that FasL participates in a positive-feedback loop to promote cell death under conditions of cellular stress.


Asunto(s)
Caspasas/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas/metabolismo , Androstadienos/farmacología , Apoptosis/efectos de los fármacos , Caspasa 3 , Caspasa 9 , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , Activación Enzimática , Proteína Ligando Fas , Proteína Forkhead Box O1 , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos , Células Jurkat , Glicoproteínas de Membrana/antagonistas & inhibidores , Glicoproteínas de Membrana/genética , Modelos Biológicos , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/metabolismo , Wortmanina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA