Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Int J Cancer ; 130(3): 532-43, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21351097

RESUMEN

The PI3 kinase/Akt pathway is commonly deregulated in human cancers, functioning in such processes as proliferation, glucose metabolism, survival and motility. We have previously described a novel function for one of the Akt isoforms (Akt3) in primary endothelial cells: the control of VEGF-induced mitochondrial biogenesis. We sought to determine if Akt3 played a similar role in carcinoma cells. Because the PI3 kinase/Akt pathway has been strongly implicated as a key regulator in ovarian carcinoma, we tested the role of Akt3 in this tumor type. Silencing of Akt3 by shRNA did not cause an overt reduction in mitochondrial gene expression in a series of PTEN positive ovarian cancer cells. Rather, we find that blockade of Akt3, results in smaller, less vascularized tumors in a xenograft mouse model that is correlated with a reduction in VEGF expression. We find that blockade of Akt3, but not Akt1, results in a reduction in VEGF secretion and retention of VEGF protein in the endoplasmic reticulum (ER). The reduction in secretion under conditions of Akt3 blockade is, at least in part, due to the down regulation of the resident golgi protein and reported tumor cell marker, RCAS1. Conversely, over-expression of Akt3 results in an increase in RCAS1 expression and in VEGF secretion. Silencing of RCAS1 using siRNA inhibits VEGF secretion. These findings suggest an important role for Akt3 in the regulation of RCAS1 and VEGF secretion in ovarian cancer cells.


Asunto(s)
Neovascularización Patológica , Neoplasias Ováricas/enzimología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Antígenos de Neoplasias/metabolismo , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Retículo Endoplásmico/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Humanos , Ratones , Ratones SCID , Mitocondrias/genética , Mitocondrias/metabolismo , Neovascularización Patológica/genética , Neoplasias Ováricas/irrigación sanguínea , Neoplasias Ováricas/genética , Fosfohidrolasa PTEN/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Transducción de Señal , Carga Tumoral/genética , Ensayos Antitumor por Modelo de Xenoinjerto
2.
J Vasc Res ; 49(2): 89-100, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22249024

RESUMEN

BACKGROUND: Poly-N-acetyl glucosamine nanofibers derived from a marine diatom have been used to increase cutaneous wound healing. These nanofibers exert their activity by specifically activating integrins, which makes them a useful tool for dissecting integrin-mediated pathways. We have shown that short-fiber poly-N-acetyl glucosamine nanofiber (sNAG) treatment of endothelial cells results in increased cell motility and metabolic rate in the absence of increased cell proliferation. RESULTS: Using a Seahorse Bioanalyzer to measure oxygen consumption in real time, we show that sNAG treatment increases oxygen consumption rates, correlated with an integrin-dependent activation of Akt1. Akt1 activation leads to an increase in the expression of the transcriptional coactivator, peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α). This is not due to increased mitochondrial biogenesis, but is associated with an increase in the expression of pyruvate dehydrogenase kinase 4 (PDK4), suggesting regulation of fatty acid oxidation. Blockade of fatty acid oxidation with etomoxir, an O-carnitine palmitoyltransferase-1 inhibitor, blocks the sNAG-dependent increased oxygen consumption. (3)H-palmitate uptake experiments indicate a PDK4-dependent increase in fatty acid oxidation, which is required for nanofiber-induced cell motility. CONCLUSIONS: Our findings imply a linear pathway whereby an integrin-dependent activation of Akt1 leads to increased PGC-1α and PDK4 expression resulting in increased energy production by fatty acid oxidation.


Asunto(s)
Acetilglucosamina/farmacología , Ácidos Grasos/metabolismo , Proteínas de Choque Térmico/biosíntesis , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factores de Transcripción/biosíntesis , Carnitina O-Palmitoiltransferasa/antagonistas & inhibidores , Compuestos Epoxi/farmacología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Nanofibras , Oxidación-Reducción , PPAR gamma/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Proteínas Quinasas/biosíntesis , Proteínas Serina-Treonina Quinasas/metabolismo , Regulación hacia Arriba
3.
FASEB J ; 22(9): 3264-75, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18524868

RESUMEN

The growth factor, vascular endothelial growth factor (VEGF), induces angiogenesis and promotes endothelial cell (EC) proliferation. Affymetrix gene array analyses show that VEGF stimulates the expression of a cluster of nuclear-encoded mitochondrial genes, suggesting a role for VEGF in the regulation of mitochondrial biogenesis. We show that the serine threonine kinase Akt3 specifically links VEGF to mitochondrial biogenesis. A direct comparison of Akt1 vs. Akt3 gene silencing was performed in ECs and has uncovered a discrete role for Akt3 in the control of mitochondrial biogenesis. Silencing of Akt3, but not Akt1, results in a decrease in mitochondrial gene expression and mtDNA content. Nuclear-encoded mitochondrial gene transcripts are also found to decrease when Akt3 expression is silenced. Concurrent with these changes in mitochondrial gene expression, lower O(2) consumption was observed. VEGF stimulation of the major mitochondrial import protein TOM70 is also blocked by Akt3 inhibition. In support of a role for Akt3 in the regulation of mitochondrial biogenesis, Akt3 silencing results in the cytoplasmic accumulation of the master regulator of mitochondrial biogenesis, PGC-1alpha, and a reduction in known PGC-1alpha target genes. Finally, a subtle but significant, abnormal mitochondrial phenotype is observed in the brain tissue of AKT3 knockout mice. These results suggest that Akt3 is important in coordinating mitochondrial biogenesis with growth factor-induced increases in cellular energy demands.


Asunto(s)
Mitocondrias/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología , Factor A de Crecimiento Endotelial Vascular/fisiología , Animales , Células Cultivadas , Endotelio Vascular/citología , Proteínas de Choque Térmico/metabolismo , Humanos , Ratones , Ratones Noqueados , Mitocondrias/efectos de los fármacos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Factores de Transcripción/metabolismo
4.
J Vasc Res ; 45(3): 222-32, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18097146

RESUMEN

Poly-N-acetyl glucosamine (pGlcNAc) nanofiber-derived materials effectively achieve hemostasis during surgical procedures. Treatment of cutaneous wounds with pGlcNAc in a diabetic mouse animal model causes marked increases in cell proliferation and angiogenesis. We sought to understand the effect of the pGlcNAc fibers on primary endothelial cells (EC) in culture and found that pGlcNAc induces EC motility. Cell motility induced by pGlcNAc fibers is blocked by antibodies directed against alphaVbeta3 and alpha5beta1 integrins, both known to play important roles in the regulation of EC motility, in vitroand in vivo. pGlcNAc treatment activates mitogen-activated protein kinase and increases Ets1, vascular endothelial growth factor (VEGF) and interleukin 1 (IL-1) expression. pGlcNAc activity is not secondary to its induction of VEGF; inhibition of the VEGF receptor does not inhibit the pGlcNAc-induced expression of Ets1 nor does pGlcNAc cause the activation of VEGF receptor. Both dominant negative and RNA interference inhibition of Ets1 blocks pGlcNAc-induced EC motility. Antibody blockade of integrin results in the inhibition of pGlcNAc-induced Ets1 expression. These findings support the hypothesis that pGlcNAc fibers induce integrin activation which results in the regulation of EC motility and thus in angiogenesis via a pathway dependent on the Ets1 transcription factor and demonstrate that Ets1 is a downstream mediator of integrin activation.


Asunto(s)
Acetilglucosamina/farmacología , Movimiento Celular/efectos de los fármacos , Células Endoteliales/fisiología , Integrinas/fisiología , Nanoestructuras , Neovascularización Fisiológica/efectos de los fármacos , Proteína Proto-Oncogénica c-ets-1/fisiología , Movimiento Celular/fisiología , Células Cultivadas , Activación Enzimática , Humanos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neovascularización Fisiológica/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo
5.
PLoS One ; 6(4): e18996, 2011 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-21559496

RESUMEN

BACKGROUND: Treatment of cutaneous wounds with poly-N-acetyl-glucosamine nanofibers (sNAG) results in increased kinetics of wound closure in diabetic animal models, which is due in part to increased expression of several cytokines, growth factors, and innate immune activation. Defensins are also important for wound healing and anti-microbial activities. Therefore, we tested whether sNAG nanofibers induce defensin expression resulting in bacterial clearance. METHODOLOGY: The role of sNAG in defensin expression was examined using immunofluoresence microscopy, pharmacological inhibition, and shRNA knockdown in vitro. The ability of sNAG treatment to induce defensin expression and bacterial clearance in WT and AKT1-/- mice was carried out using immunofluoresent microscopy and tissue gram staining. Neutralization, using an antibody directed against ß-defensin 3, was utilized to determine if the antimicrobial properties of sNAG are dependent on the induction of defensin expression. CONCLUSIONS/FINDINGS: sNAG treatment causes increased expression of both α- and ß-type defensins in endothelial cells and ß-type defensins in keratinocytes. Pharmacological inhibition and shRNA knockdown implicates Akt1 in sNAG-dependent defensin expression in vitro, an activity also shown in an in vivo wound healing model. Importantly, sNAG treatment results in increased kinetics of wound closure in wild type animals. sNAG treatment decreases bacterial infection of cutaneous wounds infected with Staphylococcus aureus in wild type control animals but not in similarly treated Akt1 null animals. Furthermore, sNAG treatment of S. aureus infected wounds show an increased expression of ß-defensin 3 which is required for sNAG-dependent bacterial clearance. Our findings suggest that Akt1 is involved in the regulation of defensin expression and the innate immune response important for bacterial clearance. Moreover, these findings support the use of sNAG nanofibers as a novel method for enhancing wound closure while simultaneously decreasing wound infection.


Asunto(s)
Acetilglucosamina/química , Nanofibras/química , Proteínas Proto-Oncogénicas c-akt/metabolismo , Cicatrización de Heridas , Animales , Antiinfecciosos/farmacología , Citocinas/metabolismo , Células Endoteliales/citología , Queratinocitos/citología , Cinética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Fluorescente/métodos , ARN Interferente Pequeño/metabolismo , beta-Defensinas/metabolismo
6.
Exp Cell Res ; 312(7): 1164-73, 2006 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-16527273

RESUMEN

Akt kinase is an important downstream effector of VEGF in primary endothelial cells (EC), promoting angiogenesis by increased cellular survival, motility and tubulogenesis. Akt1 is the founding member of a family of serine threonine kinases thought to have overlapping function. We sought to determine if other Akt family members were also regulated by VEGF in EC. We show that treatment of EC with the angiogenic inducers VEGF or sphingosine-1-phosphate (S1P) results in an increased stabilization of Akt3 mRNA, concurrent with a PI3 kinase-dependent, Akt1-independent increase in both the protein and its phosphorylation. Given the similarity of Akt3 regulation by VEGF and S1P, the sensitivity of VEGF stimulation to the Gi-protein uncoupling reagent, pertussis toxin was tested and shows that VEGF stimulation requires Gi-protein signaling. We show that the VEGF stimulates the expression of Edg3/S1P3 (S1P3) and that expression of this Gi-protein-coupled receptor is both sufficient and necessary for the expression of Akt3. Blockade of a single isoform does not overtly affect cellular function, whereas inhibition of both kinases results in an increase in apoptosis and a down-regulation of cyclin D3. These results suggest a model whereby extracellular cues maintain total Akt kinase levels through the regulation of specific isoform expression providing a fail-safe mechanism to maintain necessary levels of Akt kinase activity.


Asunto(s)
Células Endoteliales/metabolismo , Lisofosfolípidos/metabolismo , Proteínas Proto-Oncogénicas c-akt/biosíntesis , Receptores de Lisoesfingolípidos/fisiología , Esfingosina/análogos & derivados , Factor A de Crecimiento Endotelial Vascular/fisiología , Células Cultivadas , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Proteínas de Unión al GTP/fisiología , Humanos , Isoenzimas/biosíntesis , Isoenzimas/genética , Lisofosfolípidos/fisiología , Proteínas Proto-Oncogénicas c-akt/genética , Estabilidad del ARN , ARN Mensajero/metabolismo , Esfingosina/metabolismo , Esfingosina/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA