Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Carcinogenesis ; 32(4): 488-95, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21262926

RESUMEN

Oncogenic Pim-1 kinase is upregulated in multiple solid cancers, including human pancreatic ductal adenocarcinoma (PDAC), a highly lethal disease with few useful treatment options. Pim-1 is also transcriptionally induced upon oncogenic K-Ras-mediated transformation of the human pancreatic ductal epithelial (HPDE) cell model of PDAC. Given the near ubiquitous presence of mutant K-Ras in PDAC and its critical role in this disease, we wished to study the effects of oncogenic K-Ras signaling on Pim-1 expression, as well as the role of Pim-1 in growth transformation of PDAC cells. Pim-1 protein levels were upregulated in both PDAC cell lines and patient tumor tissues. Furthermore, ectopic oncogenic K-Ras increased Pim-1 expression in human pancreatic nestin-expressing (HPNE) cells, a distinct immortalized cell model of PDAC. Conversely, shRNA-mediated suppression of oncogenic K-Ras decreased Pim-1 protein in PDAC cell lines. These results indicate that oncogenic K-Ras regulates Pim-1 expression. The kinase activity of Pim-1 is constitutively active. Accordingly, shRNA-mediated suppression of Pim-1 in K-Ras-dependent PDAC cell lines decreased Pim-1 activity, as measured by decreased phosphorylation of the pro-apoptotic protein Bad and increased expression of the cyclin-dependent kinase inhibitor p27Kip1. Biological consequences of inhibiting Pim-1 expression included decreases in both anchorage-dependent and -independent cell growth, invasion through Matrigel and radioresistance as measured by standard clonogenic assays. These results indicate that Pim-1 is required for PDAC cell growth, invasion and radioresistance downstream of oncogenic K-Ras. Overall, our studies help to elucidate the role of Pim-1 in PDAC growth transformation and validate Pim-1 kinase as a potential molecular marker for mutated K-Ras activity.


Asunto(s)
Adenocarcinoma/patología , Carcinoma Ductal Pancreático/patología , Neoplasias Pancreáticas/patología , Proteínas Proto-Oncogénicas c-pim-1/fisiología , Proteínas Proto-Oncogénicas/fisiología , Tolerancia a Radiación , Transducción de Señal/fisiología , Proteínas ras/fisiología , Adenocarcinoma/radioterapia , Carcinoma Ductal Pancreático/radioterapia , Línea Celular Tumoral , Proliferación Celular , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/análisis , Humanos , Invasividad Neoplásica , Neoplasias Pancreáticas/radioterapia , Fosforilación , Proteínas Proto-Oncogénicas c-pim-1/análisis , Proteínas Proto-Oncogénicas p21(ras) , Proteína Letal Asociada a bcl/metabolismo
2.
Tetrahedron ; 66(50): 9577-9581, 2010 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-22973068

RESUMEN

The synthesis of 4-chloro-N-(naphthalen-1-ylmethyl)-5-(3-(piperazin-1-yl)phenoxy)thiophene-2-sulfonamide (B-355252) using a MW-assisted nucleophilic aromatic substitution (S(N)Ar) reaction will be discussed. Utilization of this method allowed for the rapid generation of B-355252 heteroaryl ether core structure in the presence of cesium carbonate in dimethylformamide or tripotassium phosphate in N-methyl-2-pyrrolidone in 94% yield. Evaluation of B-355252 enhancement of nerve growth factor's ability to stimulate neurite outgrowths was determined using NS-1 cells.

3.
Toxicol Sci ; 73(2): 348-61, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12700400

RESUMEN

Uncouplers of oxidative phosphorylation have relevance to bioenergetics and obesity. The mechanisms of action of chemical uncouplers of oxidative phosphorylation on biological systems were evaluated using differential gene expression. The transcriptional response in human rhabdomyosarcoma cell line (RD), was elucidated following treatment with carbonyl cyanide p-(trifluoromethoxy) phenylhydrazone (FCCP), a classical uncoupling agent. Changes in mitochondrial membrane potential were used as the biological dosimeter. There was an increase in membrane depolarization with increasing concentrations of FCCP. The concentration at 75% uncoupling (20 microM) was chosen to study gene expression changes, using cDNA-based large-scale differential gene expression (LSDGE) platforms. At the above concentration, subtle light microscopic and clear gene expression changes were observed at 1, 2, and 10 h. Statistically significant transcriptional changes were largely associated with protein synthesis, cell cycle regulation, cytoskeletal proteins, energy metabolism, apoptosis, and inflammatory mediators. Bromodeoxyuridine (BrdU) and propidium iodide (PI) assays revealed cell cycle arrest to occur in the G1 and S phases. There was a significant initial decrease in the intracellular adenosine triphosphate (ATP) concentrations. The following seven genes were selected as potential molecular markers for chemical uncouplers: seryl-tRNA synthetase (Ser-tRS), glutamine-hydrolyzing asparagine synthetase (Glut-HAS), mitochondrial bifunctional methylenetetrahydrofolate dehydrogenase (Mit BMD), mitochondrial heat shock 10-kDa protein (Mit HSP 10), proliferating cyclic nuclear antigen (PCNA), cytoplasmic beta-actin (Act B), and growth arrest and DNA damage-inducible protein 153 (GADD153). Transcriptional changes of all seven genes were later confirmed with reverse transcription-polymerase chain reaction (RT-PCR). These results suggest that gene expression changes may provide a sensitive indicator of uncoupling in response to chemical exposure.


Asunto(s)
Carbonil Cianuro p-Trifluorometoxifenil Hidrazona/toxicidad , Perfilación de la Expresión Génica , Transcripción Genética/efectos de los fármacos , Desacopladores/toxicidad , Adenosina Trifosfato/metabolismo , Carbonil Cianuro p-Trifluorometoxifenil Hidrazona/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Metabolismo Energético/genética , Femenino , Marcadores Genéticos/genética , Humanos , Membranas Intracelulares/efectos de los fármacos , Membranas Intracelulares/metabolismo , Potenciales de la Membrana/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Músculos/efectos de los fármacos , Músculos/metabolismo , Músculos/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/metabolismo , ARN Neoplásico , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Rabdomiosarcoma/genética , Rabdomiosarcoma/metabolismo , Rabdomiosarcoma/patología
4.
PLoS One ; 9(12): e114133, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25437437

RESUMEN

Adipose stem cell (ASC) differentiation is necessary for the proper maintenance and function of adipose tissue. The procurement and characterization of multipotent ASCs has enabled investigation into the molecular determinants driving human adipogenesis. Here, the transcription factor MYC was identified as a significant regulator of ASC differentiation. Expression of MYC transcript and protein was found to accumulate during the initial course of differentiation. Loss-of-function analysis using siRNA mediated knockdown of MYC demonstrated inhibition of hormonally stimulated adipogenesis. MYC exhibited an early and sustained expression pattern that preceded down regulation of key suppressor genes, as well as induction of transcriptional and functional effectors. Glucocorticoid stimulation was identified as a necessary component for MYC induction and was found to impact adipogenesis in a concentration-dependent manner. Global gene expression analysis of MYC knockdown in ASC enriched for functional pathways related to cell adhesion, cytoskeletal remodeling, and transcriptional components of adipogenesis. These results identify a functional role for MYC in promotion of multipotent ASC to the adipogenic lineage.


Asunto(s)
Adipocitos/citología , Adipogénesis , Tejido Adiposo/citología , Células Madre Adultas/citología , Proteínas Proto-Oncogénicas c-myc/genética , Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Adulto , Células Madre Adultas/metabolismo , Diferenciación Celular , Células Cultivadas , Femenino , Regulación del Desarrollo de la Expresión Génica , Humanos , Persona de Mediana Edad , Proteínas Proto-Oncogénicas c-myc/metabolismo , Interferencia de ARN
5.
Cancer Biol Ther ; 14(6): 492-501, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23760491

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is a lethal cancer with a 5-year survival rate of only 6%. Although the cytosine analog gemcitabine is the drug commonly used to treat PDAC, chemoresistance unfortunately renders the drug ineffective. Thus, strategies that can decrease this resistance will be essential for improving the dismal outcome of patients suffering from this disease. We previously observed that oncogenic Pim-1 kinase was aberrantly expressed in PDAC tissues and cell lines and was responsible for radioresistance. Furthermore, members of the Pim family have been shown to reduce the efficacy of chemotherapeutic drugs in cancer. Therefore, we attempted to evaluate the role of Pim-3 in chemoresistance of PDAC cells. We were able to confirm upregulation of the Pim-3 oncogene in PDAC tissues and cell lines versus normal samples. Biological consequences of inhibiting Pim-3 expression with shRNA-mediated suppression included decreases in anchorage-dependent growth, invasion through Matrigel and chemoresistance to gemcitabine as measured by caspase-3 activity. Additionally, we were able to demonstrate that Pim-1 and Pim-3 play overlapping but non-identical roles as it relates to gemcitabine sensitivity of pancreatic cancer cells. To further support the role of Pim-3 suppression in sensitizing PDAC cells to gemcitabine, we used the pharmacological Pim kinase inhibitor SGI-1776. Treatment of PDAC cells with SGI-1776 resulted in decreased phosphorylation of the proapoptotic protein Bad and cell cycle changes. When SGI-1776 was combined with gemcitabine, there was a greater decrease in cell viability in the PDAC cells versus cells treated with either of the drugs separately. These results suggest combining drug therapies that inhibit Pim kinases, such as Pim-3, with chemotherapeutic agents, to aid in decreasing chemoresistance in pancreatic cancer.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Carcinoma Ductal Pancreático/enzimología , Desoxicitidina/análogos & derivados , Neoplasias Pancreáticas/enzimología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Apoptosis/efectos de los fármacos , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Proliferación Celular , Desoxicitidina/farmacología , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Imidazoles/farmacología , Invasividad Neoplásica , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/patología , Fosforilación , Procesamiento Proteico-Postraduccional , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/genética , Piridazinas/farmacología , ARN Interferente Pequeño , Proteína Letal Asociada a bcl/metabolismo , Gemcitabina
6.
PPAR Res ; 2011: 179454, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22013433

RESUMEN

Growing evidence indicates that PPARγ agonists, including rosiglitazone (RSG), induce adipose mitochondrial biogenesis. By systematically analyzing mitochondrial gene expression in two common murine adipocyte models, the current study aimed to further establish the direct role of RSG and capture temporal changes in gene transcription. Microarray profiling revealed that in fully differentiated 3T3-L1 and C3H/10T1/2 adipocytes treated with RSG or DMSO vehicle for 1, 2, 4, 7, 24, and 48 hrs, RSG overwhelmingly increased mitochondrial gene transcripts time dependently. The timing of the increases was consistent with the cascade of organelle biogenesis, that is, initiated by induction of transcription factor(s), followed by increases in the biosynthesis machinery, and then by increases in functional components. The transcriptional increases were further validated by increased mitochondrial staining, citrate synthase activity, and O(2) consumption, and were found to be associated with increased adiponectin secretion. The work provided further insight on the mechanism of PPARγ-induced mitochondrial biogenesis in differentiated adipocytes.

7.
Curr Chem Genomics ; 4: 74-83, 2010 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-21347208

RESUMEN

Neurite outgrowth assays are the most common phenotypic screen to assess chemical effects on neuronal cells. Current automated assays involve expensive equipment, lengthy sample preparation and handling, costly reagents and slow rates of data acquisition and analysis. We have developed a high throughput screen (HTS) for neurite outgrowth using a robust neuronal cell model coupled to fast and inexpensive visualization methods, reduced data volume and rapid data analysis. Neuroscreen-1 (NS-1) cell, a subclone of PC12, possessing rapid growth and enhanced sensitivity to NGF was used as a model neuron. This method reduces preparation time by using cells expressing GFP or native cells stained with HCS CellMask(™) Red in a multiplexed 30 min fixation and staining step. A 2x2 camera binning process reduced both image data files and analysis times by 75% and 60% respectively, compared to current protocols. In addition, eliminating autofocus steps during montage generation reduced data collection time. Pharmacological profiles for stimulation and inhibition of neurite outgrowth by NGF and SU6656 were comparable to current standard method utilizing immunofluorescence detection of tubulin. Potentiation of NGF-induced neurite outgrowth by members of a 1,120-member Prestwick compound library as assayed using this method identified six molecules, including etoposide, isoflupredone acetate, fludrocortisone acetate, thioguanosine, oxyphenbutazone and gibberellic acid, that more than doubled the neurite mass primed by 2 ng/ml NGF. This simple procedure represents an important routine approach in high throughput screening of large chemical libraries using the neurite outgrowth phenotype as a measure of the effects of chemical molecules on neuronal cells.

8.
Int J Toxicol ; 25(1): 41-7, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16510356

RESUMEN

Multiple methods currently exist for the assessment of peroxisome proliferation, including gene expression, enzyme activity, immunolabeling coupled with image analysis, and electron microscopy. This study describes a novel flow cytometric method to efficiently quantify peroxisome proliferation in cells from frozen livers. Frozen livers from cynomolgus monkeys treated with ciprofibrate at doses of 0, 3, 30, 150, and 400 mg/kg/day for 15 days were mechanically disaggregated using an automated dispersion method. The resulting cell suspensions were labeled using an allophycocyanin (APC)-conjugated antibody directed against peroxisomal membrane protein 70 (PMP70). Statistically significant increases in mean fluorescence intensity were observed from animals dosed at 30, 150, and 400 mg/kg/day compared to control. Parallel comparisons using electron microscopy and immunofluorescence microscopy suggest that flow cytometry may be an alternative to electron microscopy in determinations of peroxisome proliferation. Flow cytometric analysis of freshly isolated hepatocytes and frozen liver from rats treated with fenofibrate at 200 mg/kg/day for 10 days showed the flow cytometric method could detect peroxisome proliferation in both species. The research described here demonstrates the feasibility of applying flow cytometry for the detection of peroxisome proliferation.


Asunto(s)
Ácido Clofíbrico/análogos & derivados , Fenofibrato/toxicidad , Citometría de Flujo/métodos , Hígado/efectos de los fármacos , Macaca fascicularis , Proliferadores de Peroxisomas/toxicidad , Peroxisomas/efectos de los fármacos , Animales , Separación Celular/métodos , Ácido Clofíbrico/toxicidad , Criopreservación , Relación Dosis-Respuesta a Droga , Estudios de Factibilidad , Ácidos Fíbricos , Hepatocitos/efectos de los fármacos , Hepatocitos/patología , Hígado/patología , Masculino , Microscopía Electrónica de Transmisión , Peroxisomas/ultraestructura , Ratas , Ratas Sprague-Dawley
9.
Cancer Immunol Immunother ; 52(7): 429-37, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12695858

RESUMEN

Ep-CAM antigen expression was shown to vary by phase across the cell cycle. Following pretreatment of various adenocarcinoma cells in culture with clinically relevant concentrations of vinorelbine tartrate (Navelbine) or paclitaxel (Taxol), cell surface expression of Ep-CAM antigen increased by two- to ten-fold compared to that of untreated control cells and was associated with arrest of cell cycle progression and accumulation of cells in the S and G2/M phases. We demonstrated that increases in cell surface antigen expression resulted in improved biological effectiveness of the targeting antibody as measured in vitro by antibody-dependent cellular cytotoxicity and in vivo by enhanced antibody targeting to Ep-CAM-expressing xenografts in mice pretreated with Navelbine. No effect on cell cycle progression or Ep-CAM antigen expression was seen with human interferon-alpha and interferon-gamma, agents that increase gene expression of various tumor and normal antigens and may upregulate some antigens. Thus, the upregulation of cell surface Ep-CAM expression following pretreatment with G2/M blockers is through a novel mechanism involving residence time of the antigen on the cell surface. This significant increase in Ep-CAM expression appears to be tumor-specific since we saw no increase in antigen expression on normal epithelial cells. Studies to reveal relative internalization rates suggest that the increase in cell surface expression of Ep-CAM following pretreatment with G2/M blockers is a consequence of an inhibition of normal cycles of antigen endocytosis and expression on the cell surface. The present work provides a mechanism for the improved clinical efficacy of therapeutic antibodies used in combination with traditional cell cycle-specific chemotherapeutic drugs.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antígenos de Neoplasias/metabolismo , Antineoplásicos Fitogénicos/farmacología , Biomarcadores de Tumor/metabolismo , Moléculas de Adhesión Celular/metabolismo , Paclitaxel/farmacología , Vinblastina/análogos & derivados , Vinblastina/farmacología , Adenocarcinoma/metabolismo , Animales , Supervivencia Celular/efectos de los fármacos , Cromo/metabolismo , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/metabolismo , Molécula de Adhesión Celular Epitelial , Femenino , Fase G2/efectos de los fármacos , Humanos , Técnicas In Vitro , Lutecio/química , Lutecio/farmacocinética , Masculino , Ratones , Ratones Desnudos , Mitosis/efectos de los fármacos , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/metabolismo , Células Tumorales Cultivadas , Vinorelbina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA